Next Article in Journal
Direct Synthesis of Phosphonates and α-Amino-phosphonates from 1,3-Benzoxazines
Next Article in Special Issue
Rare Acetogenins with Anti-Inflammatory Effect from the Red Alga Laurencia obtusa
Previous Article in Journal
Surface Enhanced Raman Spectroscopy for In-Field Detection of Pesticides: A Test on Dimethoate Residues in Water and on Olive Leaves
Previous Article in Special Issue
Erubescensoic Acid, a New Polyketide and a Xanthonopyrone SPF-3059-26 from the Culture of the Marine Sponge-Associated Fungus Penicillium erubescens KUFA 0220 and Antibacterial Activity Evaluation of Some of Its Constituents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Review on the Phytochemistry, Medicinal Properties and Pharmacological Activities of 15 Selected Myanmar Medicinal Plants

1
Department of Chemistry, Mandalay University, Mandalay 100103, Myanmar
2
Department of Chemistry, Kalay University, Kalay 03044, Sagaing Region, Myanmar
3
Departamento de Química y Ciencias Exactas, Universidad Técnica Particular de Loja (UTPL), San Cayetano Alto, Loja 11 01 608, Ecuador
*
Author to whom correspondence should be addressed.
Molecules 2019, 24(2), 293; https://doi.org/10.3390/molecules24020293
Submission received: 4 December 2018 / Revised: 9 January 2019 / Accepted: 10 January 2019 / Published: 15 January 2019
(This article belongs to the Collection Bioactive Compounds)

Abstract

:
Medicinal plants are a reservoir of biologically active compounds with therapeutic properties that over time have been reported and used by diverse groups of people for treatment of various diseases. This review covers 15 selected medicinal plants distributed in Myanmar, including Dalbergia cultrata, Eriosema chinense, Erythrina suberosa, Millettia pendula, Sesbania grandiflora, Tadehagi triquetrum, Andrographis echioides, Barleria cristata, Justicia gendarussa, Premna integrifolia, Vitex trifolia, Acacia pennata, Cassia auriculata, Croton oblongifolius and Glycomis pentaphylla. Investigation of the phytochemical constituents, biological and pharmacological activities of the selected medicinal plants is reported. This study aims at providing a collection of publications on the species of selected medicinal plants in Myanmar along with a critical review of the literature data. As a country, Myanmar appears to be a source of traditional drugs that have not yet been scientifically investigated. This review will be support for further investigations on the pharmacological activity of medicinal plant species in Myanmar.

Graphical Abstract

1. Introduction

Natural products, especially those derived from plants, have been used to help mankind sustain human health since the dawn of medicine. Traditional medicine has been in existence since time immemorial and has been well accepted and utilized by the people throughout history. Since ancient times, plants have been an exemplary source of medicines. Plant-derived medicinal products have attracted the attention of scientists around the world for many years due to their minimum side effects and positive effects on human health.
In the pharmaceutical landscape, plants with a long history of use in ethnomedicine can be a rich source of substances for the treatment of various ailments and infectious diseases. Medicinal plants are considered a repository of numerous types of bioactive compounds possessing varied therapeutic properties. The vast array of therapeutic effects associated with medicinal plants includes anti-inflammatory, antiviral, antitumor, antimalarial, and analgesic properties.
According to the World Health Organization (WHO), a variety of drugs are obtained from different medicinal plants and about 80% of the world’s developing population depends on traditional medicines for their primary health care needs [1]. Myanmar (Figure 1) has abundant plant resources and Myanmar peoples have used their own traditional medicines to maintain their health and treat various ailments, including malaria, diarrhea and fever over millennia of history [2].
We previously reported [3] the traditional uses, chemical constituents, the published the biological and pharmacological evidences of 25 medicinal plants from Kachin State, in Northern Myanmar. In Kachin State, the climatic conditions vary considerably, from warm to humid in the lowlands, to extremely cold in the highlands, and the mountains in the far north are snowcapped. The geographical and climate conditions of the central region in Myanmar are quite different from those of Kachin State. That region has a generally harsh climate and is extremely dry. Because of the geographical and climate differences the chemical constituents of the plants from that region have different medicinal uses, biological and pharmacological activities. Therefore, in this review, it is out intention to provide a complete account of the phytochemistry, medicinal and pharmacological uses of a few plants which grow in the central region of Myanmar, which is the tropical area of Myanmar. The traditional healers in central Myanmar use a lot of species of plants that serve as regular sources of medicine and there is a need to preserve the traditional knowledge on the uses of these medicinal plants.
A total of 15 medicinal plants, which belong to seven families and 15 genera were recorded in this review. The results are summarized in Table 1, which provides the following information for each species: scientific name, botanical family and local name. The most represented families are the Fabaceae with six species, followed by Acanthaceae (three species), Verbenaceae (two species), and Mimosaceae, Casealpiniaceae, Euphorbiaceae and Rutaceae were represented with one species each. The primary objective of this study is to present a database of the indigenous knowledge on medicinal plants used for various diseases among the local traditional healers of Myanmar, to describe the chemical constituents and to summarize the published biological and pharmacological evidence. The focus of this review is to provide information on the phytochemical constituents reported in the literature and published biological and pharmaceutical activities of fifteen medicinal plants from Myanmar.
Until now, few sources of information are available for some medicinal plants in the literature, and the sources of information in the literature make evident that additional new investigations and scientific reports are necessary.

2. Phytochemistry, Medicinal Properties and Ethnopharmacology of the Selected Myanmar Medicinal Plants

2.1. Dalbergia culrata Grah. (DC)

D. cultrata, locally known as Yin-daik, is widely distributed in Myanmar [4]. It is a moderate-sized deciduous tree indigenous to the forests of Burma that belongs to the family Fabaceae (or Leguminosae) [5]. This plant genus is mainly distributed in tropical and subtropical regions, such as South Africa and India, Laos, Myanmar, Thailand and Vietnam in Asia [6,7].
The plant family is known to contain isoflavonoids and neoflavonoids [8]. The chemical constituents from the heartwood of this plant includes 4-arylcoumarin derivatives; (S)-4-methoxy dalbergione (1) and stevenin (2), a p-hydroquinone derivative, dalbergin (3), and diphenylpropene derivatives 3,3-diphenylprop-1-ene (4) and 3-(5-acetoxy-2,4-dimethoxyphenyl)-3-(3′-acetoxyphenyl) prop-1-ene (5) [9]. Ito isolated cinnamoylphenol type compounds, dalberatin A (6) and dalberatin B (7) (Figure 2) from the stem bark of D. cultrate which showed remarkably potent tumor-promotion inhibitory activity in vitro with the IC50 value of 212 and 303 (mol ratio/32 pmol TPA), suggesting that they might be valuable as potential cancer chemopreventive agents [10].

2.2. Eriosema chinense Vogel.

E. chinense (family Fabaceae) is locally known in Myanmar as pike-san-gale. It is a small plant and two members of its genus are found in Myanmar. In Myanmar traditional medicine, a decoction of the seeds is used for scrofula, diarrhoea, to treat leucorrhoea and menstrual derangements. In 2009, Sutthivaiyakit’s group isolated eight prenylated flavonoids, khonklonginols A–H (815), five flavonoids, lupinifolinol (16), dehydrolupinifolinol (17), flemichin D (18), eriosemaone A (19), lupinifolin (20), and one lignin, yangambin, from the hexane and dichloromethane extracts of the roots of E. chinense [11] (Figure 3).
From the hexane, dichloromethane and methanol extract of the roots of E. chinense, seven flavonoids, trans-p-coumaric acid ester together with another twelve compounds were identified as 3-epilupinifolinol (21), 3-epikhonklonginol C (22), 2′-hydroxylupinifolinol (23), 2,5,2,4-tetrahydroxy-6″,6″-dimethylpyrano(2″,3″:7,6)-8-(3‴,3‴-dimethylallyl) flavone (24), (2R,3R,2‴R)-3,5,2‴-trihydroxy-4-methoxy-6″,6″-dimethylpyrano(2″,3″:7,6)-8-(3‴-methylbut-3‴-enyl) flavanone (25), 2‴,3‴-epoxy-khonklonginol A (26), 6,7-dimethoxy-5,2′,4′-trihydroxyisoflavone (27), octaeicosanyl-trans-p-coumarate, khonklonginol A (8), lupinifolinol (16), flemichin D (18), 7-O-methyltectorigenin, tectorigenin (28), genistein (29), kaempferol (30), 2′,4′,5,7-tetrahydroxy-6-methoxyisoflavone, kaempferol-7-O-β-d-glucopyranoside (31), genistein-7-O-β-d-glucopyranoside, genistin, astragalin (32) and cajanol [12].
Prasad evaluated the antidiarrhoeal activity of the roots of ethanol extract, its fractions (chloroform, ethyl acetate and hexane) and lupinifolin isolated from the chloroform fraction of the roots. Lupinifolin showed moderate antidiarrhoel activity compared to the bioactive chloroform fraction [13].
A prenylated flavanone, eriosematin E (33) was isolated from the roots of E. chinense and significant antidiarrhoel activity results were observed, with the maximum effective dose of eriosematin E (10 mg/kg p.o.) in inhibiting peristaltic index (small intestinal transit) and reducing intestinal fluid volume of castor oil induced and PGE 2 induced enteropooling models. In addition, eriosematin E restored all the alterations in biochemical parameters such as nitric oxide, protein, DNA, superoxide dismutase, catalase and lipid peroxidation and also significantly recovered Na+ and K+ loss from body [14]. The cytotoxic inhibitory activity of khonklonginol A (8), khonklonginol B (9) and lupinifolinol (16) showed IC50 values of 3.1, 3.8 and 1.73 μg/mL against cell lung NCI-H187 cell line. Dehydrolupinifolinol (17), flemichin D (18), eriosemaone A (19) and lupinifolin (20) were showed the antimycobacterial against Mycobacterium tuberculosis H37Ra with MIC values, in turn, 25, 12.5, 12.5 and 12.5 μg/mL [11]. Khonklonginol A (8), lupinifolinol (16), flemichin D (18), 3-epi-lupinifolinal (21), 2′-hydoxylupinifolinol (23), 3,5,2,4-tetrahydroxy-6″,6″-dimethylpyrano (2″,3″:7,6)-8-(3‴,3‴-dimethylallyl) flavone (24), tectorigenin (28), genistein (29), kaempferol (30), kaempferol-7-O-β-d-glycopyranoside (31) and astragalin (32) showed antimicrobial and antioxidant activity [12]. Among these compounds, khonklonginol A showed the most effectively activity against Candida albicans, gram negative bacteria, Escherichia coli, Klebsiella pneumoniae and Pseudomonas aeruginosa and positive bacteria, Bacillus cereus, Enterococcus faecalis, Staphylococcus aureus, MRSA S. aureus, S. epidermidis, S. agalactiae and S. pyrogenes. A detailed description of the antimicrobial and antioxidant activities of these compounds is given in Table 2. The antidiarrhoel activity of the various extracts of the roots of E. chinense has been reported [13].

2.3. Erythrina suberosa Roxb.

E. suberosa locally known as ka-thit and widely distributed in Myanmar. It belongs to the family Fabaceae, and is an ornamental tall tree also found occasionally on hills and moist slopes of India, Pakistan, Nepal, Bhutan, Burma, Thailand and Vietnam [15]. In traditional Myanmar medicine, the bark is used for dysentery, while the leaves are reported to be used in urinary diseases and inflammation. In India, E. suberosa has been used as a very important medicinal plant for the treatment of various ailments [16]. The bark is reported to be used in preparation of medicines for dysentery, as an astringent and febrifuge, and used for anorexia, liver troubles, helminthic manifestations, inflammations, intestinal worms, obesity and as a snake-bite antidote [17].
The species of this genus are famous for different classes of compounds like alkaloids, flavonoids and terpenes. One prenylated flavanone 5,7,3′,4′-tetrahydroxy-2′-(3″-methyl but-2″-enyl)-flavanone (34) (Figure 4) isolated from the ethanol extract of the roots of this plant [18]. According to the Kumar group’s report, the stem bark of E. suberosa contains α-hydroxyerysotrine (35), 4′-methoxy licoflavanone (36), alpinumisoflavone (37) and wighteone (38) [19].
The seed powder of E. suberosa contains erythraline (39), erysodine (40), erysotrine (41) and hypaphorine [20]. Serrano’s group isolated two erythrinian alkaloids, erysodine and erysotrine, from the ethanolic extract of the flowers of E. suberosa [15]. The dichloromethane extract of the wood of E. suberosa is reported to contain erysubin A (42), erysubin B (43), erythrinin C (44), alpinumisoflavone, a wighteone metabolite, wighteone and laburnetin [21]. Dichloromethane extract of the roots of this plant have been reported to contain petrocarpans, erysubin C (45), erysubin D (46), erysubin E (47), erysubin F (48), orientanol D, bidwillon B, 2-(γ,γ-dimethylallyl)-6a-hydroxyphaseollidin, phaseollin, erythrabyssin II, cristacarpin, wighteone, bidwillon A, alpinumisoflavone and erystagallin A [22].
Serrano observed that acute p.o. treatment with the isolated alkaloids; erysodine (40) and erysotrine (41) produced anxiolytic effects in the elevated plus-maze and the light-dark transition model [15]. The anticancer potential and cytotoxic effect of some compounds from E. suberosa has been reported scientifically. Thus, Kumar studied the cytotoxic effects of 4′-methoxylicoflavanone (36) and alpinumiso-flavone (37) from E. suberosa on apoptosis in human leukemia HL-60 cells and their potential to induce cancer cell death [19]. The ethanolic extract of the leaves of E. suberosa showed antitumor activity [23]. In addition, the ethanolic extract of the stem bark of E. suberosa induced apoptosis in human promyelocytic leukemia HL-60 cells [24]. It seems that E. suberosa has the potential for being selected for future anticancer therapeutics.

2.4. Millettia pendula BENTH.

M. pendula is locally called thin-win and is widely distributed in Myanmar. M. pendula also belongs to the family Fabaceae. Its timber is produced in Myanmar and Thailand [25]. In Myanmar traditional medicine, the root of M. pendula is applied for skin diseases, allergy, urinary diseases and inflammation, based on folklore medicine. Hayashi found the pterocarpan, maackiain and isoflavans, pendulone and claussenquinone in the heartwood of M. pendula and confirmed their identity and purification through NMR [26].
In 2006, three isoflavans: millettilone A (49), 3R-claussequinone (50) and pendulone (51), a 2-arylbenzofuran—millettilone B (52)—three pterocarpans: secundiflorol I (53), 3,8-dihydroxy-9-methoxypterocarpan (54) and 3,10-dihydroxy-7,9-dimethoxypterocarpan (55), and one isoflavone, formononetin (56), were isolated from a methanol extract of the powdered M. pendula timber [25] (Figure 5).
Takahashi reported the vitro leishmanicidal activity of the methanolic timber extract of M. pendula [25]. Among the constituents of M. pendula pendulone (51), an isoflavan, showed significant leishmanicidal activity with an IC50 value of 0.07 μg/mL, while millettilone A (49, IC50 9.3 μg/mL), 3R-claussequinone (50, IC50 1.2 μg/mL) and 3,8-dihydroxy-9-methoxypterocarpan (54, IC50 2.9 μg/mL) exhibited moderate activity and secundiflorol I (53, IC50 86 μg/mL) and 3,10-dihydroxy-7,9-dimethoxypterocarpan (55, IC50 77 μg/mL) displayed weak leishmanicidal activity in vitro [25]. There are few chemical constituents and biological studies of this plant reported in the scientific literature, therefore, biologically active new chemical components from this plant might yet be found.

2.5. Sesbania grandiflora (L.) Poir.

S. grandiflora is a small, erect, fast-growing, and sparsely branched cultivated tree belonging to the family Fabaceae. It’s locally known as paukpan-phyu in Myanmar. In folk medicine, the juice of the leaves and flowers is used as a popular remedy for catarrh, and headache. The bark of S. grandiflora is very astringent and the bitter bark is considered a tonic. Decoctions of leaves and flowers is used to treat leucorrhoea and vomiting of blood. The bark of S. grandiflora is used as an astringent and treatment of small pox, ulcers in the mouth and the alimentary canal, infant stomach disorders and scabies. The juice of the leaves of S. grandiflora has been reportedly used in the treatment of bronchitis, cough, vomiting, wounds ulcers, diarrhea, and dysentery. The flowers have reported antimicrobial activity. Powdered roots of this plant are mixed in water and applied externally as a poultice or rub for rheumatic swelling [27,28].
According to qualitative analysis, the methanol extract of the leaves of S. grandiflora is reported to contain alkaloids, glycosides, steroids, terpenoids and tannins. 3,4,5-Trimethoxyphenol, erucic acid, 2-furancarboxaldehyde, vitamin E acetate, 4-methyloxazole, palmitic acid, 9-hexadecenol, and dioctyl ester are the major compounds in the leaves of S. grandiflora by GC-MS analysis [29]. Three isoflavonoids—isovestitol (57), medicarpin (58) and sativan (59)—and one lupane type triterpene, betulinic acid (60, Figure 6) were isolated from the methanol extract of the roots of S. grandiflora [30]. High contents of quercetin, myricetin and kaempferol were identified in a methanolic extract of the leaves [31] and a novel protein fraction was isolated from the fresh flowers, which displayed chemopreventive effects [32]. Isovestitol (57), medicarpin (58) and sativan (59) all displayed significant antitubercular activity, with MIC values of 50 μg/mL each, but betulinic acid (60) showed weak activity against Mycobacterium tuberculosis H37Rv with a MIC value of 100 μg/mL in vitro. The crude extract of the root of S. grandiflora exhibited a moderate antitubercular activity [27].
Remesh has reported that S. grandiflora has protective effects against cigarette smoke-induced oxidative damage in the liver and kidney of rats and protected the lungs from oxidative damage through its antioxidant potential [33,34]. Doddola reported the antiurolithiatic and antioxidant activities of the leaf juice of S. grandiflora [35]. The ethanol extract of the leaves and flowers of S. grandiflora exhibited anticancer activity against an Ehrlich ascites carcinoma tumor model [36]. The leaves of S. grandiflora showed a significant protection against erythromycin estolate-induced toxicity [27], antimicrobial [37], anticonvulsant and anxiolytic activity [38]. The alcoholic extract of S. grandiflora flowers at 250 and 500 mg/kg exhibited significant antidiabetic activity (p < 0.01) in alloxan- induced diabetic rats [39]. The S. grandiflora flowers displayed significant hepatoprotective [40] and antimicrobial activity [28]. Silver nanoparticles synthesized using the leaf extract of this plant showed not only excellent antibacterial activity against clinically isolated multi-drug resistant human pathogens [41], but also antifungal activity against some selected microbial pathogens [42].

2.6. Tadehagi triquetrum (L.) H. Ohashi.

T. triquetrum (synonyms: Desmodium triquetrum (L.) DC.) belongs to the family Fabaceae, which is widely distributed throughout the tropical, subtropical and Pacific regions of the world. This shrub is commonly known as lauk-thay or shwe-gu-than-hlet in Myanmar and is distributed in the regions of Chin, Kachin, Kayin, Mandalay, Sagaing, Shan and Yangon [4] and the southern area of Yunan Province in China [43]. An infusion of the roots is taken to treat kidney complaints, and an infusion of the leaves is drunk for stomach discomfort in Myanmar. A daily glass of a decoction of the roots of this plant is considered beneficial for chronic coughs and tuberculosis.
In the whole plant of T. triquetrum, Xiang et al., in 2005 and Zhang et al., in 2011 reported prenylated isoflavones—triquetrumone A (61), triquetrumone B (62), and triquetrumone C (63)—a prenylated biisoflavanone-(R)—triquetrumone D (64)—together with cyclokievitone, yukovanol, aromadendrin, kaempferol, astragalin, 2-O-methyl-l-chiro-inositol, galactitol, p-hydroxycinnamic acid, ursolic acid, betulinic acid, β-sitosterol, daucosterol, stigmasterol, stigmasta-5,22-dien-3-O-β-d-glucopyranoside, saccharose and docosanoic acid [43], and isoflavanones: triquetrumones E–H (6568) [44]. From the aerial parts of T. triquetrum, one lignin—tadehaginosin (69)—along with 3,4-dihydro-4-(4′-hydroxyphenyl)-5,7-dihydroxycoumarin (70) [45], and ten phenylpropanoid glucosides-tadehaginosides A–J (7180), along with tadehaginoside (81) were isolated [46] (Figure 7).
In Myanmar, local people use T. triquetru leaves to cover local fish pastes to prevent the development of fly larvae. A study in Myanmar proved that the aqueous extract of T. triquetrum has bactericidal activity on some pathogenic bacteria [47]. Moe et al. reported that the leaves of T. triquetrum were used by tuberculosis patients in Myanmar. The family members of patients were also familiar with this plant. In the central region of Myanmar, the herbs are available in the forest fringes but sometimes available in local markets of rural areas [48]. The extract of T. triquetrum and tadehagenoside proved to display hypoglycemic activity [49], with in vivo immunoprotection effects because of its antioxygenic properties [50], and anti-inflammatory activity [51]. Moreover, tadehaginosin (69) and 3,4-dihydro-4-(4′-hydroxyphenyl)-5,7-dihydroxycoumarin (70) from T. triquetrum showed hypoglycemic activity in vitro on HepG2 cells [45]. The phenylpropanoid glucosides-tadehaginosides C–J (7380) and tadehaginoside (81) isolated from T. triquetrum showed the potential to be developed into antidiabetic compounds [46], providing the scientific evidence needed for developing new chemical candidates in treating diabetes. In Myanmar, there are more and more diabetes patients nowadays, and based on the literature review, the plant T. triquetrum could be a very useful and valuable plant for treating diabetes patients.

2.7. Andrographis echioides Nees.

A. echioides is an annual medicinal plant of Acanthaceae family and the plants are seen mostly in dry places, such as South India, Sri Lanka and South Asian countries [52]. In Myanmar, it is distributed in the central region—Mandalay and Sagaing—and known by the common name se-ga-gyi-hmawe-tu. There is no traditional knowledge of A. echioides use in Myanmar till now based on folk medicine, but in Indian traditional medicine, the leaf juice of A. echioides is used to cure fevers [53]. The leaves of A. echioides boiled with coconut oil is applied to decrease the loss and graying of hair [54].
From the whole plant of A. echioides, Jayaprakasam’s group characterized flavanone- dihydroechioidinin (82) (Figure 8) [55], flavones—echioidinin (83) [56], echioidin (84), skullcapflavone I 2′-O-methyl ether (85)—flavone glucosides—skullcapflavone I 2′-O-glucoside (86) [57,58], and echioidinin 5-O-β-d-glucoside (87) [59] and a chalcone glucoside, androechin (88). 2′-Oxygenated flavonoids and phenyl glycosides from the whole plant of A. echioides include androgechosides A–B (89,90), androechiosides A–B (91,92) together with 2′,6′-dihydroxy-acetophenone 2′-O-β-d-glucopyranoside (93), pinostrobin (94), andrographidine C (95), dihydroechioidinin, tectochrysin 5-glucoside (96), methyl salicylate glucoside, 7,8-dimethoxy-5-hydroxyflavone (97), 5,7,8-trimethoxyflavone (98), skullcapflavone I 2′-methyl ether, acetophenone-2-O-β-d-glucopyranoside, androechin, skullcapflavone I 2′-O-β-d-glucopyranoside, tectochrysin (99), negletein 6-O-β-d-glucopyranoside, andrographidine E, 4-hydroxy-3-methoxy-trans-cinnamic acid methyl ester, 4-hydroxybenzaldehyde, 4-hydroxy-trans-cinnamic acid methyl ester, O-coumaric acid, 2,6-dihydrobenzoic acid, 132-hydroxy-(132-R)-phaeophytin, (E)-phytyl epoxide, phytol, phytene 1,2-diol, (+)-dehydrovomifoliol, 3β-hydroxy-5α,6α-epoxy-7-megastigmen-9-one, β-sitosterol, β-sitosteryl-3-O-β-glucopyranoside, squalene, 1H-indole-3-carbaldehyde, and loliolide were reported by Shen’s group [58]. Moreover, a variety of chemical compounds isolated from the petroleum ether, ethyl acetate and methanol extracts of A. echioides were analysed by GC-MS [60].
A flavanone, dihydroechioidinin (82), and 5,7,8-trimethoxyflavone (98) isolated from A. echioides are reported to possess potent anti-inflammatory activity, with IC50 values of 37.6 ± 1.2 μM and 39.1 ± 1.3 μM, respectively, and to suppress NO inhibition activity [58].
Basu reported not only the hepatoprotective and antioxidant activity of methanolic extract of the aerial parts of A. echioides against acetaminophen-induced hepatotoxicity in rats, but also the anti-inflammatory, analgestic and antipyretic activities of the ether, chloroform and ethyl acetate extracts of this plant [61,62]. The chloroform extract of the leaves of A. echioides exhibited significant diuretic activity [63]. The ethanol extract from the leaves of A. echioides showed antiulcer activity [64] and antifungal activity in vitro [65]. Rajkumar et al., in 2012 reported that the synergistic larvicidal activity of the leaf extracts of A. echioides against larvae of Aedes aegypti L [66]. Thus, we can say that A. echioides is a good source of antioxidant and antimicrobial compounds. The ethanolic extract of A. echioides has notable antibacterial activity against both Gram positive and negative bacteria in vitro [67] and the hydroalcoholic extract of this plant showed antimicrobial activity [54]. The methanol extract of A. echioides displayed potent antioxidant activity IC50 values on the range of 1.14 ± 0.06 to 1.15 ± 0.45 mg/mL, as well as antibacterial activity (16 ± 1.527 mm) than the aqueous extract of this plant (20 ± 1.527 mm) [68]. While the phytochemical constituents and biological activities of the extracts of A. echioides have been reported, however, the pharmacological action of pure compounds from this plant has not been investigated yet, and therefore might represent an interesting research topic.

2.8. Barleria cristata L.

B. cristata or leik-tha-ywe-pya is a perennial shrub, belonging to the family Acanthaceae. It is found widely in subtropical Himalaya, Sikkim, Kashi Hills, and southern India at a height of 1350 meter [69]. The bitter juice of the leaves of B. cristata is used as a diaphoretic and expectorant for serious catarrhal infections in the central region of Myanmar. In addition, an infusion of the roots and leaves is applied to boils and sores to reduce swelling. B. cristata is traditionally used for the treatment of a variety of diseases including anemia, toothache and coughs in Indian traditional medicine. Roots and leaves are used to reduce swelling and inflammation [70].
Phytochemical screening by HPTLC analysis revealed that the presence of amino acids, carbohydrates, flavonoids, glycosides, saponins, steroids, terpenoids, tannins, alkaloids and phenolic compounds in the plant extract [71]. In 2005, three phenylethanoid glycosides, desrhamnosyl acteoside (100), acteoside (101) and poliumoside (102), were isolated from the callus cultures of B. cristata [72], followed by two phenolic compounds, p-coumaric acid (103) and α-tocopherol (104), two flavonoidal compounds, luteoline (105) and 7-methoxyluteoline (106), and iridodial glycosides, barlerin (107) and schanshiside methyl ester (108) from the leaves of B. cristata (Figure 9) [73]. Besides these compounds, researchers have found 4-hydroxy-trans-cinnamate derivatives and oleanolic acid (109) in this plant [74]. Biological investigations of this plant showed anti-inflammatory [69,75], and antioxidant properties [76,77]. There are few phytochemistry and biological studies of this species; therefore, it seems to be interesting for further investigation of new lead products.

2.9. Justicia gendarussa Burm F.

J. gendarussa (Family: Acanthaceae) is a shade-loving, quick-growing, evergreen plant mostly found in moist areas. In Myanmar, it is commonly known as pha-wa-net and distributed in Bago, Kachin, Mandalay, Shan, Taninthayi and Yangon. J. gendarussa is native to China and is distributed widely across India, in tropical and subtropical areas of Asia, Sri Lanka and Malaysia [78]. In traditional medicinal systems, different parts of J. gendarussa have been used in a variety of diseases. The leaves and tender shoots are diaphoretic and used for chronic rheumatism. The juice of the fresh leaves is dropped into the ear for treating earache and an oil prepared from the leaves is used topically to treat oedema caused by beriberi. The plant has been used by the native medical practitioners and tribes to treat various ailments including liver disorders, tumours, inflammation and skin diseases [79].
Two diphyllin glycosides were obtained from the stems and the barks of J. gendarussa; justiprocumins A–B (110,111) [80] (Figure 10). Souza et al. isolated four alkaloids, brazoides A–D (112115), squalene, β-sitosterol and lupeol from the ethanol extract of leaves of J. gendarussa [81].
Patentiflorin A (116) from J. gendarussa displayed anti-HIV activity against a broad spectrum of HIV strains with IC50 values in the range of 24–37 nM, higher than the clinically used first anti-HIV drug, zidovudine AZT (IC50 77–95 nM) [82]. In addition, justiprocumin B (111) showed potent activity against a broad spectrum of HIV strains with IC50 values in the range of 15–21 nM (AZT, IC50 77–95 nM), nevirapine-resistant isolate HIV-1N119 with an IC50 value of 495 nM and AZT-resistant isolate HIV-11617-1 with (IC50 185 nM). The compound also showed potent inhibitory activity against the NRTI-resistant isolate HIV-1 of the analogue AZT as well as the NNRTI-resistant isolate (HIV-1N) of the analogue nevaripine [80]. J. gendarussa had been reported to have a variety of pharmacological actions like hepatoprotective [83], antioxidant [84,85], anti-inflammatory and analgesic [85], antiangiogenic [86], anti-anxiety [87], anti-arthritic [88], anti-cancer [89,90], antibacterial [91], antifungal [92] and anthelmintic [93] effects based on folk medicine uses. The ethyl acetate soluble fraction isolated from the methanolic extract of the roots of J. gendarussa possessed anti-inflammatory activity in the carrageenan-induced paw edema model [94]. Scientific evidence for these pharmacological actions has not yet been found. It seems that J. gendarussa has the potential for being selected for further clinical studies. Moreover, relatively few chemical constituents of this plant are studied, hence it might be of interest to search for new bioactive compounds in its extracts.

2.10. Premna integrifolia L.

The plant P. integrifoia belongs to the family Verbenaceae, widely distributed in Vietnam, Laos and Cambodia [95]. In Myanmar, it is locally known by the names taung-tangyi or kywe-thwe and widely distributed in Mandalay, Rakhine and Taninthayi [4]. Traditionally, roots and stem barks are used as a laxative, carminative, and stomachic. Decoctions of the whole plant are used for fevers, rheumatism and neuralgia. The roots are used in the treatment of diabetes, inflammation, swelling, bronchitis, dyspepsia, liver disorders, piles, constipation and fever [96]. The decoction of the roots is used in gonorrhea and during convalescence from fever [97]. Two alkaloids, premnine and ganiarine, are isolated from the stem bark of P. integrifolia [97]. It is widely used by the traditional practitioners as a cardiotonic, antibiotic, anti-coagulant, stomachic, carminative, hepatoprotective and antitumor agent [98].
Two iridoid glycosides along with other ten compounds were isolated by Hang et al., in 2008 from the flowers: premnacorymboside A (117) (Figure 11), 10-O-trans-p-methoxycinnamoylcatalpol (119), and verbascoside, from the leaves: premnacorymboside B (118), 10-O-trans-p-methoxy-cinnamoylcatapol, scutellarioside II (120), premnaodoroside A (121), 1-O-trans-p-coumaroyl-α-l-rhamnopyranoside, hexyl glucoside, 4-hydroxy-2-methoxybenzaldehyde, and 4-hydroxy-benzaldehyde, from the stem bark of P. integrifolia: premnaodoroside A, scutellarioside II, quercetin-3-rutinoside, and leonuriside A [99]. Three diterpenoids, 1β,3α,8β-trihydroxy-pimara-15-ene (122), 6α,11,12,16-tetrahydroxy-7-oxoabieta-8,11,13-triene (123) and 2α,19-dihydroxypimara-7,15-diene (124) were identified from the root bark of P. integrifolia [100]. Major iridoid glycosides, 10-O-trans-p-methoxycinnamoylcatapol, 4″-hydroxy-E-globularinin (125) and premnosidic acid (126) [101] and two furofuran lignans, premnadimer (127) and 4β-hydroxyasarinin-1-O-β-glucopyranoside (128) together with other nine compounds were isolated from the stem bark of P. integrifolia and structurally elucidated by Yadav et al. [102].
Premnadimer (127), 4β-hydroxyasarinin-1-O-β-glucopyranoside (128), and iridoids 10-O-trans-p-methoxycinnamoylcatapol (119, IC50 0.37 μM/mL), 4″-hydroxy-E-globularinin (125, IC50 0.37 μM/mL), premnosidic acid (126) and 10-O-trans-p-coumaroyl-6-O-α-l-rhamnopyranosylcatapol showed antioxidant activity in the DPPH and NO free radical scavenging assays. Compound 126 and 10-O-trans-p-coumaroyl-6-O-α-l-rhamnopyranosylcatapol displayed a maximum ferric reducing ability in the FRAP assay [101]. Twenty-nine compounds representing 94.81% of the leaf oil were analysed by GC-MS. Among these, phytol (27.25%), α-humulene (14.21%), spathulenol (12.12%), 1-octen-3-ol (8.21%), eugenol (6.69%), phenylethyl alcohol (5.81%) and caryophyllene oxide (2.6%) were the major compounds. The major components of the oil from P. integrifolia and extracts of the leaves of this plant displayed great antibacterial activity potential against some bacterial strains such as Sarcina lutea, Bacillus subtilis, Escherichia coli, Pseudomonas sp., Klebsiella pneumoniae and Xanthomonas campestris [103]. In addition, the methanolic extract of P. integrifolia bark was reported to have antidiabetic activity along with antioxidant activity in vitro [104] and anti-inflammatory activity [105]. The traditional uses of the plant still need to be investigated scientifically.

2.11. Vitex trifolia L.

V. trifolia belongs to the Verbenaceae family, a small shrub, is distributed widely in Southeast Asia, Micronesia, Australia, East Africa [106], and Yunan, Guangxi, and Guangdong provinces in mainland China [107]. It is commonly known in Myanmar as kyaung-ban. In Myanmar, dried fruit powder is orally taken with honey in a dose of 5–10 gm for diarrhoeal diseases. For menstrual disorders and urinary disorders, indigestion, the dried powder is orally taken with roasted common salt and warmed water in the same dose as above. Leaves is used as a curry ingredient. Root powder is slurried with alcohol and externally used for muscle cramps.
From the fruits of V. trifolia, Ono reported isolation of an abietane-type diterpene, vitetrifolin A (129) (Figure 12), two labdane-type diterpenes, vitetrifolins B (130) and C (131), and diterpenes, rotundifuran (132), dihydrosolidagenone (133) and abietatriene 3β-ol (134) [108]. Moreover, Ono’s group also reported halimane-type diterpenes, vitetrifolins D–G (135138) from the fruits of this plant [109]. Fruits of V. trifolia were found to contain five labdane-type diterpenes, vitexilactione, (rel 5S,6R,8R,9R,10S)-6-acetoxy-9-hydroxy-13(14)-landen-16,15-olide, rotundifuran, vitetrifolin and vitetrifolin E [110] and flavonoids, persicogenin (139), artemetin (140), luteolin (141), penduletin (142), vitexicarpin (143) and chrysosplenol-D (144) [111]. A series of labdane-type diterpenoids, vitextrifolins A–G (145151), together with rotundifuran, vitetrifolin H, vitexilactone, vitexilactone B, deacetylvitexilactone, viteagnusin I, previtexilactone and negundol were isolated from the fruits of V. trifolia [112]. Additionally, an iridoid agnuside (152) was found in the leaves of the same species [113].
Negundoside (153), agnuside (152) and 6-p-hydroxybenzoyl mussaenosidic acid (154) isolated from the aerial parts of V. trifolia showed significant antioxidant activity with IC50 values of 9.96, 9.81 and 10.31 μg, respectively in DPPH and 16.25, 12.90 and 13.51 μg in NO radical scavenging assays [114]. The ethanol extract of the leaves of V. trifolia displayed free radical scavenging activity [113]. The methyl-p-hydroxybenzoate (155), which is reported from V. trifolia possessed potential mosquito larvicidal activity with the LC50 values of 5.77 ppm against Culex quinquefasciatus and 4.74 ppm against Aedes aegypti [115].
Essential oils of fresh leaves of V. trifolia, which was extracted by hydro-distillation in a Clevenger’s apparatus, were evaluated against Vth instar larvae of Spilosoma obliqua, for insect growth regulatory activity [116]. According to Matsui et al. the leaves of V. trifolia showed the significant inhibitory activity on interleukin (IL)-1-β, IL-6 and inducible nitric oxide synthase (iNOS) mRNA synthesis and have a little effect on tumor necrosis factor (TNF) [117] and it acts on inflammatory activity related gene expression by inhibiting the NF-κB action [118]. The labdane-type diterpenoids [110] and flavonoids from the fruits of this plant [111] displayed anti-proliferative effects on cancer cells through the induction of the apoptosis and the inhibition of the cell cycle. Biological assays have been proved that the hexane and dichloromethane extracts of this plant are highly cytotoxic against several cancer cell lines [119]. The biological activities of V. trifolia have been reported scientifically, however, some traditional uses are still uninvestigated.

2.12. Acacia pennata (L.) Willd.

A. pennata, belonging to the family Mimosaceae, is a large woody prickly climber with bipinnate leaves. A. pennata (local name in Myanmar suyit or suboke-gyi) is widely distributed in regions of south and southeast Asia including Bangladesh, Butan, India, Sri Lanka, southwest China and Thailand [120]. In Myanmar, barks of A. pennata have been used in the treatment of asthma and bronchitis. It is reported to have some medicinal value. The leaf juice when mixed with milk is given to infants for indigestion. The A. pennata plant is used as an antiseptic for burning urine and for curing bleeding gums. The root bark can be used as antiflatulent and to cure stomach pain. It is also used in the treatment of bronchitis, cholera and asthma. A decoction of the leaves is used for general treatment of body aches, headaches and fevers [121].
From the leaves of A. pennata, two diterpenoids, a flavonoid glycoside together with another compounds were discovered: taepeenin D (156) (Figure 13), (+)-drim-8-ene (157), quercetin 3-O-β-d-glucopyranosyl-4-O-β-d-glucopyranoside (158), labdanolic acid and 8,15-labdanediol [122], quercetin 4′-O-α-l-rhamnopyranosyl-3-O-β-d-allopyranoside (159), apigenin 6-C-[2″-O-(E)-feruloyl-β-d-glucopyranosyl]-8-C-β-glucopyranoside (160), together with isorhamnetin 3-O-α-l-rhamno-pyranoside (161), kaempferol 3-O-α-l-rhamnopyranosyl-(1→4)-β-d-glucopyranoside (162), and isovitexin (163) [123]. From the aerial parts of A. pennata, a species distributed in Popa Mountain National Park (Mandalay, Myanmar), five flavonoid glycosides; (2R,3S)-3,5,7-trihydroxyflavan-3-O-α-l-rhamnopyranoside (164), (2S)-5,7-dihydroxyflavan-7-O-β-d-glucopyranoside-(4α→8)-epi-afzelechin-3-O-gallate (165), (2R)-4′,7-dihydroxyflavan-(4α→8)-(2R,3S)-3,5,7-trihydroxyflavan-3-O-α-l-rhamnopyranoside (166), 5,7-dihydroxyflavone-6-C-β-boivinopyranosyl-7-O-β-d-gluco-pyranoside (167), and 5,7-dihydroxyflavone 7-O-β-d-glucopyranosyl-8-C-β-boivinopyranoside (168), along with other compounds, quercetin-3-O-β-d-glucopyranoside (169), quercetin-3-O-α-l-rhamnopyranoside (170), chrysin-7-O-β-d-glucopyraniside (171), kaempferol 3-O-α-l-rhamnopyranoside (172), koaburanin (173) and pinocembrin-7-O-β-d-glucopyranoside (174) were isolated [124].
Taepeenin D (156), (+)-drim-8-ene (157) and quercetin 3-O-β-d-glucopyranosyl-4-O-β-d-glucopyranoside (158) showed cytotoxic activity against human pancreatic (PANC1) cancer with IC50 values of 3.2 μM, 15.1 μM and 26.6 μM, respectively, and prostate cancer (DU145) cells, with IC50 values of 3.4 μM, 23.2 μM and 30.0 μM, respectively, and no toxic effects on normal cells. Taepeenin D (156), (+)-drim-8-ene (157) and quercetin 3-O-β-d-glucopyranosyl-4-O-β-d-glucopyranoside (158), which possessed Hedgehog/GLi-mediated transcriptional activity, showing IC50 values of 1.6, 13.5 and 10.5 μM [122]. Biological activities of A. pennata were reported to include anti-parasitic [117] anti-nociceptive and anti-inflammatory [123,125], and antioxidant effects [126]. The phytochemical constituents and biological activities of A. pennata have been reported, however, further pharmacological studies are still needed to scientifically prove the basis of their actions.

2.13. Cassia auriculata Linn.

C. auriculata belongs to the family Caesalpiniaceae, which is an ethno-botanically important shrub with attractive yellow flowers. It is cultivated in Myanmar and commonly known as peik-thin-khet. The ethanolic extract of the roots of C. auriculata displayed the nephroprotective activity in cisplatin and gentamicin induced renal injury in male albino rats [127]. The plant has been reported to possess antipyretic, hepatoprotective, antidiabetic, antiperoxidative and antihyperglyceamic and microbicidal activity [128,129]. The flowers are used to treat urinary discharges, nocturnal emissions, diabetes and throat irritation. The leaves are anthelmintic and good for ulcers, skin diseases and leprosy [130].
Nakamura reported a benzocoumarin glycoside, avaraside I (175, Figure 14), along with other compounds such as av170araol I (176), luteolin, kaempferol, quercetin (177), myricetin (178), 3-methoxy-luteolin (179), kaempferol 3-O-β-d-glucopyranoside (180), isoquercetin (181), myricetin-3-O-β-d-glucopyranoside (182), kaempferol-3-O-rutinoside (183), rutin (184), myricetin 3-O-rutinoside (185), lanceolatin B (186), pseudosemiglabrin (187), (+)-catechin, (+)-gallocatechin, (−)-epicatechin, 6-demethoxycapillarisin, 6-demethoxy-7-methylcapillarisin, (2S)-7,4-dihydroxyflavan(4β→8)-catechin (188), (2S)-7,4-dihydroxyflavan(4β→8)-gallocatechin (189), (2S)-7,4-dihydroxyflavan(4β→8)-epicatechin (190), (2S)-7,4-dihydroxyflavan(4β→8)-epigallocatechin (191), chrysophanol, emodin, physcion, roseoside, bridelionoside C, benzyl-O-β-d-apiofuranosyl (1→2)-β-d-glucopyranoside and (−)-epigallocatechin (192) from the leaves of C. auriculata [131].
The ethanolic extract of C. auriculata flowers showed antihyperlipidemic activity [132]. The aqueous extract of the leaves of C. auriculata possessed antihyperglycemic and hypolipidemic activity in streptozotocin (STZ)-induced mild diabetic and severe diabetic rats [133]. The methanol extract of the root of this plant had significant hepatoprotective activities and antitubercular drug-induced hepatotoxicity [134]. Pseudosemiglabrin (187) [inhibition %: 37.0 ± 3.7 (p < 0.01) at 30 μM], (2S)-7,4-dihydroxyflavan (4β→8)-catechin (188) [inhibition %: 33.5 ± 2.9 (p < 0.01) at 30 μM], (2S)-7,4-dihydroxyflavan (4β→8)-gallocatechin (189) [inhibition %: 28.2 ± 4.7 (p < 0.01) at 30 μM], and (−)-epigallocatechin (192) [inhibition %: 24.4 ± 3.1 (p < 0.01) at 30 μM], isolated from C. auriculata displayed hepatoprotective effects against cytotoxicity induced by D-GalM in primary cultured mouse hepatocytes [135]. C. auriculata leaf extract has a protective action against alcohol-induced oxidative stress to the cells as evidenced by the lowered tissue lipid peroxidation and elevated levels of the enzymic and non-enzymic antioxidants [135]. Polyphenols derived from the flowers of C. auriculata showed immunomodulatory effects on various immune cell compositions and their relevant functions [136]. The biological activities of chemical constituents and active principle(s) from this plant are still lacking thorough investigations.

2.14. Croton oblongifolius Roxb.

C. oblongifolius (Euphorbiaceae), is popularly known as thetyin-gyi in Mynanmar. It is a medium sized tree widely distributed in the region of Mount Popa in Myanmar. Seeds are used as diarrhea and oedema, and very useful for inflammation, either taken orally or as an external application in Myanmar. Root bark and seeds are reported to be used as purgative, liver diseases and high blood pressure. It is used in gastric ulcers; the bark is used to treat dyspepsia and the roots to treat dysentery [137].
From the leaves of C. oblongifolius, a clerodane type diterpene, nasimalun A (193) (Figure 15) was isolated [138]. Nasimalun A (193) from the leaves of C. oblongifolius displayed moderate cytotoxicity toward MOLT-3 cell line with IC50 value of 12.0 μM. In addition, this compound showed antibacterial activity showing the MIC values of 50, 12.5, and 100 μg/mL for Bacilus cereus and both Staphylococcus aureus, and Staphylococcus epidermidis respectively [139]. Clerodane type diterpenes, crovatin (194), methyl 15,16-epoxy-3,13(16),14-ent-clerodatrien-18,19-olide-17-carboxylate (195) and dimethyl 15,16-epoxy-12-oxo-3,13(16),14-ent-clerodatriene-17,18-dicarboxylate (196), nasimaluns A and B (197), levatin, (−)-hardwickiic acid, 15-hydroxy-cis-ent-cleroda-3,13(E)-diene (198) and a sesquiterpene, patchoulenone were reported from the roots of C. oblongifolius [140]. Two cleidtanthane-type diterpenoids, hydroxycleistantha-13(17),15-ene (199) and 3,4-seco-cleistantha-4(18),13(17),15-triene-3-oic acid (200) [141], four furanocembranoids 1–4 (201204) [142], (−)-ent-kuar-16-en-19-oic acid (205) [138], furoclerodane-type diterpene, croblongifolin (206), one clerodane, crovatin, eight labdane-types, nidorellol (207), 2-acetoxy-3-hydroxylabda-8(17),12(E)-14-triene (208), 3-acetoxy-2-hydroxylabda-8(17),12(E),14-triene (209), 2,3-dihydroxylabda-8(17),12(E),14-triene (210) [143], labda-7,12(E),14-triene (211), labda-7,12(E),14-triene-17-al (212), labda-7,12(E),14-triene-17-ol (213) and labda-7,12(E),14-triene-17-oic acid (214) [144], three cembranoids, neocrotocembranal (215) [145], crotocembraneic acid (216) and neocrotocembraneic acid (217) [146], one diterpene acid, (+)-isopimara-7(8),15-diene-19-oic acid [147], oblongifoliol (218) [148] were obtained from the stem bark of C. oblongifolius.
Croblongifolin (206) isolated from the stem bark of C. oblongifolius was displayed cytotoxicity towards several human tumor cell lines including HepG2 (IC50 0.35 μM), SW620 (IC50 0.47 μM), CHAGO (IC50 0.24 μM), KATO3 (IC50 0.35 μM) and BT474 (IC50 0.12 μM) [143]. Neocrotocembranal (215) was also reported cytotoxicity against P-388 cells in vitro with an IC50 6.48 μg/mL and inhibited platelet aggregation induced by thrombin [145]. (−)-ent-kuar-16-en-19-oic acid (205) was possessed the inhibition of Na+, K+-ATPase activity with an IC50 of 2.2 × 10−5 M [138]. Furanocembranoid 1 (201) proved to be cytotoxic for human tumor cell lines BT474, CHAGO, Hep-G2, KATO-3 and SW620s, with IC50 values of 7.8, 7.0, 5.6, 5.9 and 6.3 μg/mL, respectively by MTT assay. Compound (203) showed inhibitory activity against BT474 cell lines (IC50 9.6 μg/mL), CHAGO (IC50 7.1 μg/mL), Hep-G2 (IC50 5.7 μg/mL), KATO-3 (IC50 8.2 μg/mL) and SW-620s (IC50 5.6 μg/mL), while (204) inhibited the growth of the human tumor BT474 (IC50 9.6 μg/mL), CHAGO (IC50 9.3 μg/mL), Hep-G2 (IC50 6.1 μg/mL), KATO-3 (IC50 8.1 μg/mL) and SW-620s (IC50 6.0 μg/mL) cell lines [142]. In 2016, Adiga evaluated the anti-tumor activity of the methanolic extract of the stem bark of C. oblongifolius [148].
The methanolic extract of the aerial part of C. oblongifolius showed maximum antihepatotoxic activity, whereas, the other extracts (petroleum ether and acetone) showed lower activity [149]. Phytochemical constituents and biological activities of C. oblongifolius have been reported scientifically.

2.15. Glycosmis pentaphylla Correa

G. pentaphylla is a shrub or small tree, ca. 1.5–5.0 m high, and is widely distributed from India, Malaysia, and Southern China to the Philippine Islands [150]. It is commonly known as taw-shauk or obok in Myanmar and is widely distributed in Bago, Magway, Mandalay and Yangon. In traditional Myanmar medicine, juice of the leaves of G. pentaphylla is used in fevers, liver complaints and as a vermifuge. A paste of the leaves mixed with ginger is applied for eczema and skin affections. A decoction of the roots is given for facial inflammation. G. pentaphylla is a species of plants belonging to the Rutaceae family, which has been used in several countries as a traditional medicinal plant for the treatment of various ailments. It is used for the treatment of cough, rheumatism, anaemia, and jaundice. Stems and roots of this plant are used for treatment of ulcer [151].
A naphthoquinone, glycoquinone (219) and acridone alkaloid, glycocitrine-III (220) (Figure 16) [152], four hydroquinone diglycoside acyl esters, glypentosides A–C (221223) and seguinoside F (224) [153], carbazole alkaloid, 4-(7-hydroxy-3-methoxy-6-methyl-9H-carbazol-4-yl)but-3-en-2-one (225), two dimeric carbazole alkaloids, bisglybomine B (226) and biscarbalexine A (227), together with glybomine B, glycoborinine, carbalexine A, 4,8-dimethoxy-1-methyl-3-(3-methylbut-2-en-1-yl)-quinolin-2(1H)-one, 4,8-dimethoxyfuro [2,3-b]quinoline, skimmianine and arborinine [154], three phenolic glycosides, glycopentosides D–F (228230) [155] and two carbazole indole-type dimeric alkaloids, glycosmisines A (231) and B (232) [156] were isolated from the stems of G. pentalphylla. Alkaloid, glycophymine (233) and an amide, glycomide from the flowers of this species [157] and derivative of carbazole alkaloid, glycozolinine (234) was identified from the seeds of G. pentaphylla [158]. In 1966, alkaloid, skimmianine (235) and acridone alkaloids, noracronycine (236), des-N-methylacronycine (237) and des-N-methylnoracronycine (238) were isolated from the root bark of G. pentaphylla [159] and followed by carbazole (239) and 3-methylcarbazole (240) [160] from the same parts of this species in 1987. Bhattacharya obtained the carbazole alkaloids, glycozolidol (241) [161], glycozolidal (242) [162] from the roots of G. pentalphylla. One quinolone alkaloid, glycolone [163], three furanopyridine alkaloids, glypenfurans A-C, along with skimmianine, γ-fagarine, R-(+)-platydesmine, melicarpine, robustine and haplopine [164] were isolated from the leaves of the same plant. The hydroperoxyquinolone alkaloid, glycopentaphyllone, acutifolin, 3-(3′,3′-dimethylallyl)-4,8-dimethoxy-N-methylquinolin-2-one, glycocitlone C, arborine, dictamine, γ-skimmianine, 1-hydroxy-3,4-dimethoxy-10 methylacridan-9-one and arborinine were reported from the fruits of G. pentaphylla [165].
The methanolic extract of the leaves and stems of G. pentaphylla showed antimicrobial, antioxidant and cytotoxic effects [166]. The acridone alkaloid arborinine, isolated from G. pentaphylla, showed significant inhibitory activity towards the growth of crown gall tumors produced by Agrobacterium tumefaciens in a potato disc bioassay [167]. The methanolic extract of the leaves of G. pentaphylla showed hepatoprotective activity against paracetamol-induced hepatotoxicity in Swiss albino mice [168]. Streejith et al., reported in 2012 the anti-hepatocellular carcinoma cell line activity of G. pentaphylla ethanol extract in reducing the proliferation of Hep3 B cells [151]. Biscarbalexine A (227), glycosmisines A (231) and B (232) displayed significant the cytotoxic activity against human cancer A549, HepG-2 and Hun-7 cell lines, showing with an IC50 values of 56.05, 43.68 and 57.01 μM for A549 cell line, 60.06, 50.30 and 62.89 μM for HepG-2 cell line and 73.16, 30.60 and 62.87 μM for Huh-7 cell line, respectively [156]. G. pentaphylla ethanolic crude extract showed a significant antioxidant activity [169]. According to the literature, this plant seems to be a rich source of bioactive compounds. Various biological and pharmacological activities of selected medicinal plants and their products have been summarized in Table 2.

3. Conclusions

In this review, we have systematically summarized the structures and properties of 242 compounds isolated from 15 species of selected Myanmar medicinal plants. Among the 15 selected medicinal plants that have been described in this review, Erythrina suberosa might have potential as a future anticancer therapeutic based on the literature. The plants, Croton oblongifolius, Eriosema chinense, Premna integrifolia and Erythrina suberosa, their phytochemical constituents and biological activities are extensively investigated. Studies on the plants of Acacia pennata, Cassia auriculata, Glycomis pentaphylla and Tadehagi triquetrum have reported the presence of several bioactive compounds whose biological and pharmacological applications can further be investigated. The chemical and pharmacological action of Vitex trifolia have mainly emphasized on fruits, however, the chemical constituents of other parts of this plant and some traditional uses are still missing. On the other hand, the biological activities of the various extracts of Sesbania grandiflora, and Andrographis echioides have been reported scientifically but there have been a few reports on chemical constituents. There are few phytochemistry and biological studies of Dalbergia cultrata and Millettia pendula, Barleria cristata and Justicia gendarussa; Acacia pennata and Cassia auriculata, therefore, they seem to be interesting for investigation of new pharmaceuticals.
A vast reservoir of selected species remains untapped in terms of phytochemical constituents, as well as pharmacology, and this is the research gap for future studies. Further investigations of endemic plants in Myanmar and their phytochemical constituents are necessary to completely understand the molecular mechanisms of their action in vivo and in vitro and to assure the plant extracts are safe for human use.

Author Contributions

M.M.A., H.T.A., M.M.S., and C.A. share the contributions of field work, reviewed the literatures and compilation of this manuscript. All authors have read and approved the final manuscript.

Funding

M.M.A. is grateful to the Beijing University of Chemical Technology for a China Scholarship Council (CSC), Chinese Government Scholarship (2017–2018).

Conflicts of Interest

All authors declare to have no actual or potential conflict of interest.

References

  1. Fransworth, N.R.; Akerele, O.; Bingel, A.S.; Soejarto, D.D.; Guo, Z. Medicinal plants in therapy. Bull. WHO 1985, 63, 965–981. [Google Scholar] [CrossRef]
  2. Soe, K.; Ngwe, T.M. Medicinal Plants of Myanmar: Identification and Uses of Some 100 Commonly Used Species Series (1), 1st ed.; Shaung, T., Ed.; Forest Resource Environment Development and Conservation Association: Yangon, Myanmar, 2004; p. 255. [Google Scholar]
  3. Aung, H.T.; Sein, M.M.; Aye, M.M.; Thu, Z.M. A review of traditional medicinal plants from Kachin State, Northern Myanmar. Nat. Product Commun. 2016, 11, 353–364. [Google Scholar]
  4. Kress, W.J.; Defilipps, R.A.; Farr, E.; Kyi, Y.Y. A Checklist of the Trees, Shrubs, Herbs, and Climbers of Myanmar, Smithsonian Institution; The United States National Herbarium: Washington, DC, USA, 2003; Volume 45, pp. 1–590. [Google Scholar]
  5. Gamble, J.S. A Manual of Indian Timbers; Sampson Low Marston & Company: Marston, London, UK, 1922; p. 253. [Google Scholar]
  6. Takahashi, M.; Fuchino, H.; Satake, M.; Agatsuma, Y.; Sekita, S. In vitro screening of leishmanicidal activity in Myanmar timber extracts. Biol. Pharm. Bull. 2004, 27, 921–925. [Google Scholar] [CrossRef] [PubMed]
  7. Saha, S.; Shilpi, J.A.; Mondal, H.; Hossain, F.; Anisuzzman, M.; Hasan, M.M.; Cordell, G.A. Ethnomedicinal, phytochemical, and pharmacological profile of the genus Dalbergia L. (Fabaceae). Phytopharmacology 2013, 4, 291–346. [Google Scholar]
  8. Eyton, W.B.; Ollis, W.D.; Sutherland, I.O.; Gottlieb, O.R.; Magalhaes, T.; Jackman, L.M. The neoflavonoid group of natural products-I: Dalbergions- a new class of quinones. Tetrahedron 1965, 21, 2683–2696. [Google Scholar] [CrossRef]
  9. Donnelly, D.M.X.; O’reilly, J.; Thompson, J. Neoflavonoids of Dalbergia cultrata. Phytochemistry 1972, 11, 823–826. [Google Scholar] [CrossRef]
  10. Ito, C.; Itoigawa, M.; Kanematsu, T.; Runagrungsi, N.; Higashihara, H.; Tokuda, H.; Nishino, H.; Furukawa, H. New cinnamylphenols from Dalbergia species with cancer chemopreventive activity. J. Nat. Prod. 2003, 66, 1574–1577. [Google Scholar] [CrossRef]
  11. Sutthiyaiyakit, S.; Thongnak, O.; Lhinhatrakool, T.; Yodchun, O.; Srimark, R.; Dowtaisong, P.; Chuankamnerdkarn, M. Cytotoxic and antimycobacterial prenylated flavonoids from the roots of Eriosema chinense. J. Nat. Prod. 2009, 72, 1092–1096. [Google Scholar] [CrossRef]
  12. Prasad, S.K.; Laloo, D.; Kumar, M.; Hemalatha, S. Antidiarrhoeal evaluation of root extract, its bioactive fraction, and lupinifolin isolated from Eriosema chinense. Planta Med. 2013, 79, 1620–1627. [Google Scholar] [CrossRef]
  13. Prasad, S.K.; Parmar, K.M.; Danta, C.C.; Laloo, D.; Hemalatha, S. Antidiarrhoeal activity of eriosematin E isolated from the roots of Eriosema chinense Vogel. Phytomedicine 2017, 24, 127–133. [Google Scholar] [CrossRef]
  14. Thongnest, S.; Lhinhatrakool, T.; Wetprasit, N.; Sutthivaiyakit, S. Eriosema chinense: A rich source of antimicrobial and antioxidant flavonoids. Phytochemistry 2013, 96, 353–359. [Google Scholar] [CrossRef] [PubMed]
  15. Serrano, M.A.R.; Batista, A.N.-D.-L.; Bolzani, V.-D.-S.; Santos, L.-D.-A. Anxiolytic-like effects of erythrinian alkaloids from Erythrina suberosa. Quim. Nova. 2011, 34, 808–811. [Google Scholar]
  16. Chopra, R.N.; Nayar, S.L.; Chopra, I.C. Glossary of Indian Medicinal Plants; Council of scientific & Industrial Research: New Delhi, India, 1956. [Google Scholar]
  17. Gitte, T.A.; Kare, M.A.; Deshmukh, A.M. Ethno-medicinal studies on barks of some medicinal plants in Marathwada (M.S.), India-I. Recent Res. Sci. Technol. 2012, 4, 8–10. [Google Scholar]
  18. Chauha, P.; Saxena, V.K. A new prenylated flavone from Erythrina suberosa roots. Planta Med. 1987, 53, 221–222. [Google Scholar] [CrossRef] [PubMed]
  19. Kumar, S.; Pathania, A.S.; Saxena, A.K.; Vishwakarma, R.A.; Ali, A.; Bhushan, S. The anticancer potential of flavonoids isolated from the stem bark of Erythrina suberosa through induction of apoptosis and inhibition of STAT signalling pathway in human leukemia HL-60 cells. Chem.-Biol. Int. 2013, 205, 128–137. [Google Scholar] [CrossRef] [PubMed]
  20. Singh, H.; Chawla, A.S. Erythrina sp. 111: Chemical constituents of Erythrina suberosa Roxb. Seeds. J. Pharm. Sci. 1970, 59, 1179–1182. [Google Scholar] [CrossRef] [PubMed]
  21. Tanaka, H.; Tanaka, T.; Etoh, H.; Watanabe, N.; Ahmad, M.; Qurashi, I.; Khand, M.R. Two new isoflavones from Erythrina suberosa var. glabrescences. Heterocycles 1998, 48, 2661–2666. [Google Scholar] [CrossRef]
  22. Tanaka, H.; Etoh, H.; Watanabae, N.; Shimizu, H.; Ahmad, M.; Rizwani, G.H. Erysubins, C-F, four isoflavonoids from Erythrina suberosa var. glabrescencens. Phytochemistry 2001, 56, 769–773. [Google Scholar] [CrossRef]
  23. Dhar, M.L.; Dhar, M.M.; Dhawan, B.N.; Mehrotra, B.N.; Ray, C. Screening of Indian plants for biological activity: Part-I. Indian J. Exp. Biol. 1968, 6, 232–247. [Google Scholar] [PubMed]
  24. Agrawal, S.K.; Agrawal, M.; Sharma, P.R.; Gupta, B.D.; Arora, S.; Saxena, A.K. Induction of Apoptosis in Human Promyekicytic Leukemia HL60 cells by an extract from Erythrina suberosa stem bark. Nutr. Cancer 2011, 63, 802–813. [Google Scholar] [CrossRef] [PubMed]
  25. Takahashi, M.; Fuchino, H.; Sekita, S.; Satake, M.; Kiuchi, F. In vitro leishmanicidal constituents of Millettia pendula. Chem. Pharm. Bull. 2006, 54, 915–917. [Google Scholar] [CrossRef]
  26. Hayashi, Y.; Shirato, T.; Sakurai, K.; Takahashi, T. Isoflavonoids from the heartwood of Millettia pendula. Mojuzaki Gakkaishi 1978, 24, 898–901. [Google Scholar]
  27. Pari, L.; Uma, A. Protective effect of Sesbania grandiflora against erythromycin estolate induced hepatotoxicity. Therapie 2003, 58, 439–443. [Google Scholar] [CrossRef]
  28. Munde-Wagh, K.B.; Wagh, V.D.; Toshniwal, S.S.; Sonawane, B.R. Phytochemical, antimicrobial evaluation and determination of total phenolic and flavonoid contents of Sesbania grandiflora flower extract. Int. J. Pharm. Sci. 2012, 4, 229–232. [Google Scholar]
  29. Hussain, A.Z.; Kumaresan, S. GC-MS studies and phytochemical screening of Sesbania grandiflora L. J. Chem. Pharm. Res. 2014, 6, 43–47. [Google Scholar]
  30. Husan, N.; Osman, H.; Mohamad, S.; Chong, W.K.; Awang, K.; Zahariluddin, A.S.M. The chemical components of Sesbania grandiflora root and their antituberculosis activity. Pharmaceuticals 2012, 5, 882–889. [Google Scholar] [CrossRef]
  31. Mustafa, R.A.; Hamid, A.A.; Mohamed, S.; Bakar, F.A. Total phenolic compounds, flavonoids, and radical scavenging activity of 21 selected tropical plants. J. Food Sci. 2010, 75, C28–C35. [Google Scholar] [CrossRef]
  32. Laladhas, K.P.; Cheriyan, V.T.; Puliappadamba, V.T.; Bava, S.V.; Unnithan, R.G.; Vijayammal, P.L.; Anto, R.J. A novel protein fraction from Sesbania grandiflora shows potential anticancer and chemopreventive efficacy, in vitro and in vivo. J. Cell. Mol. Med. 2010, 14, 636–646. [Google Scholar]
  33. Ramesh, T.; Mahesh, R.; Begum, V.H. Effect of Sesbania grandiflora on lung antioxidant defense system in cigarette smoke exposed rats. Int. J. Biol. Chem. 2007, 1, 141–148. [Google Scholar]
  34. Ramesh, T.; Sureka, C.; Bhuvana, S.; Begum, V.H. Sesbania grandiflora diminishes oxidative stress and ameliorates antioxidant capacity in liver and kidney or rats exposed to cigarette smoke. J. Physiol. Pharmacol. 2010, 61, 467–476. [Google Scholar]
  35. Doddola, S.; Pasupulati, H.; Koganti, B.; Prasad, K.V. Evaluation of Sesbania grandiflora for antiurolithiatic and antioxidant properties. J. Nat. Med. 2008, 62, 300–307. [Google Scholar] [CrossRef]
  36. Sreelatha, S.; Padma, P.R.; Umasankari, E. Evaluation of anticancer activity of ethanol extract of Sesbania grandiflora (Agati Sesban) against Ehrlich ascited carcinoma in Swiss albino mice. J. Ethnopharmacol. 2011, 134, 984–987. [Google Scholar] [CrossRef]
  37. Kumar, N.S.; Dhanyaraj, F.S. Phytochemical analysis and antimicrobial activities of Sesbania grandiflora (L) leaf extracts. Int. J. Pharm. Sci. Rev. Res. 2016, 36, 144–148. [Google Scholar]
  38. Kasture, V.S.; Deshmukh, V.K.; Chopde, C.T. Anxiolytic and anticonvulsive activity of Sesbania grandiflora leaves in experimental animals. Phytother. Res. 2002, 16, 455–460. [Google Scholar] [CrossRef]
  39. Kumar, R.; Janadri, S.; Kumar, S.; Dhanajaya, D.R.; Swamy, S. Evaluation of antidiabetic activity of alcoholic extract of Sesbania grandiflora flower in alloxan induced diabetic rats. Asian J. Pharm. Pharmacol. 2015, 1, 21–26. [Google Scholar]
  40. Kale, I.; Khan, M.A.; Irfan, Y.; Goud, A.V. Hepatoprotective potential of ethanolic and aqueous extract of flowers of Sesbania grandiflora (Linn) induced by CCl4. Asian Pac. J. Trop. Biomed. 2012, 2, S670–S679. [Google Scholar] [CrossRef]
  41. Das, J.; Das, M.P.; Velusamy, P. Sesbania grandiflora leaf extract mediated green synthesis of antibacterial silver nanoparticles against selected human pathogens. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2013, 104, 265–270. [Google Scholar] [CrossRef]
  42. Ajitha, B.; Reddy, Y.A.K.; Rajesh, K.M.; Reddy, P.S. Sesbania grandiflora leaf extract assisted green synthesis of silver nanoparticles: Antimicrobial activity. Mater. Today Proc. 2013, 3, 1977–1984. [Google Scholar] [CrossRef]
  43. Xiang, W.; Li, R.-T.; Mao, Y.-L.; Zhang, H.-J.; Li, S.-H.; Song, Q.-S.; Sun, H.-D. Four new prenylated isoflanonoids in Tagehagi triquetrum. J. Agric. Food. Chem. 2005, 53, 267–271. [Google Scholar] [CrossRef]
  44. Zhang, R.-T.; Cheng, G.-G.; Feng, T.; Cai, X.-H.; Luo, X.-D. Four new isoflavanones from Tadehagi triquetrum. Nat. Prod. Bioprospect. 2011, 1, 121–123. [Google Scholar] [CrossRef]
  45. Wu, J.; Zhang, C.Y.; Zhang, T.; Zhao, D.; An, N.; Li, Y.; Zhu, N.; Wang, S.; Chen, F.; Zhang, X. Four new prenylated isoflavonoids in Tadehagi triquetrum. Nat. Prod. Res. 2015, 29, 1–5. [Google Scholar]
  46. Zhang, X.; Chen, C.; Li, Y.; Chen, D.; Dong, L.; Na, W.; Wu, C.; Zhang, J.; Li, Y. Tadehaginosides, A-J, phenylpropanoid glucosides from Tadehagi triquetrum, enhance glucose uptake via the upregulation of PPARy and GLUT-4 in C2C12 myotubes. J. Nat. Prod. 2016, 79, 1249–1258. [Google Scholar] [CrossRef]
  47. Lwin, H.S.; Tu, M. Effect of Desmodium triquetrum extract on some pathogenic bacteria. Union Burma J. Life Sci. 1968, 1, 66–70. [Google Scholar]
  48. Moe, S.; Naing, K.S.; Htay, M.N.N. Herbal medicines used by tuberculosis patients in Myanmar. Euro. J. Med. Plants 2018, 22, 1–10. [Google Scholar] [CrossRef]
  49. Wang, Y.Q.; Feng, B.M.; Tang, L.; Shi, L.Y.; Shi, H.F. Research of Tadehagi triquetrum (L.) DC. J. Dalian Univ. 2007, 28, 49–51. [Google Scholar]
  50. Zhou, X.D.; Shi, L.Y.; Yu, D.Y.; Shi, H.B.; Miao, Y.B.; Tang, L.; Feng, B.M.; Wang, Y.Q. Effect of active part extracted from Tadehagi triquetrum (Linn.) Ohashi on type I allergy induced by immunoglobulin E. Central South. Pharm. 2011, 9, 35–38. [Google Scholar]
  51. Tang, A.; Chen, X.; Lu, Q.; Zheng, N.; Wei, Y.; Wu, X. Antihepatotoxic effect of tadehaginoside, extracted from Tadehagi triquetrum (L.), against CCl4-lesioned rats through activating the Nrf2 signaling pathway and attenuating the inflammatory response. Inflammation 2014, 37, 1006–1014. [Google Scholar] [CrossRef]
  52. Gamble, J.S. Flora of the Presidency of Madras; Botanical Survey of India: Calcutta, India, 1956; Volume 2, p. 1051. [Google Scholar]
  53. Kirtikar, K.R.; Basu, B.D. Indian Medicinal Plants; Bishen Singh Mahendra Pal Singh: New Delhi, India, 1975; Volume 3, pp. 1884–1886. [Google Scholar]
  54. Kanchana, G.; Nirubama, K.; Rubalakshmi, G. Phytochemical screening and antimicrobial activity of Andrographis echioides (L.) Nees-an indigenous medicinal plant. World J. Pharm. Pharm. Sci. 2014, 3, 702–710. [Google Scholar]
  55. Govindachar, T.R.; Parthasarathy, P.C.; Pai, B.R.; Subramania, P.S. Chemical examination of Andrographis echioides-I: Structure and synthesis of echioidinin. Tetrahedron 1965, 21, 2633–2640. [Google Scholar] [CrossRef]
  56. Govindachar, T.R.; Parthasarathy, P.C.; Pai, B.R.; Subramania, P.S. Chemical examination of Andrographis echioides-II: Structure and synthesis of echioidin. Tetrahedron 1965, 21, 3715–3720. [Google Scholar] [CrossRef]
  57. Jayaprakasam, B.; Damu, A.G.; Gunasekar, D.; Blond, A.; Bodo, B. Dihydroechioidinin, a flavanone from Andrographis echioides. Phytochemistry 1999, 52, 935–937. [Google Scholar] [CrossRef]
  58. Shen, D.-Y.; Juang, S.-H.; Kuo, P.-C.; Huang, G.-J.; Chan, Y.-Y.; Damu, A.G.; Wu, T.-S. Chemical constituents from Andrographis echioides and their anti-inflammatory activity. Int. J. Mol. Sci. 2013, 14, 496–514. [Google Scholar] [CrossRef]
  59. Jayaprakasam, B.; Gunasekar, D.; Rao, K.V.; Blond, A.; Bodo, B. Androechin, a new chalcone glucoside from Andrographis echioides. J. Asian Nat. Prod. Res. 2001, 3, 43–48. [Google Scholar] [CrossRef]
  60. Mathivanan, D.; Suseem, S.R. Identified bioactive constituents on leaf of Andrographis echioides grown on Vellore District, Tamil Nadu. Der Pharm. Lett. 2015, 7, 138–147. [Google Scholar]
  61. Basu, S.K.; Rupeshkumar, M.; Kavitha, K. Hepatoprotective and antioxidant effect of Andrographis echioides N. against acetaminophen induced hepatotoxicity in rats. J. Biol. Sci. 2009, 9, 351–356. [Google Scholar]
  62. Basu, S.K.; Rupeshkumar, M.; Kavitha, K. Studies on the anti-inflammatory, analgestic and antipyretic properties of Andrographic echioides Nees. Int. J. Pharmacol. 2009, 5, 251–256. [Google Scholar]
  63. Murthy, J.R.; Rajaraman, M.; Meera, R. Phytochemical constituents and diuretic activity of leaf extracts of Androgarphis echioides-l-Nees. Int. J. Pharm. Chem. Sci. 2012, 1, 1659–1665. [Google Scholar]
  64. Raja, R.R.; Jeevanreddy, K. Pharmacognostical phytochemical and anti-ulcer activity of Andrographis echioides (Acanthaceae). J. Pharm. Phytochem. 2014, 3, 39–49. [Google Scholar]
  65. Punnagai, K.; Chellathai, D.D.; Karthik, V.P.; Josephine, I.G. Evaluation of antifungal activity of ethanolic extract of Andrographis echioides—An in vitro study. Int. J. Pharm. Bio. Sci. 2016, 7, 6–10. [Google Scholar]
  66. Rajkumar, S.; Jebanesan, A.; Nagarajan, R. Synergistic effect of Andrographis echioides and Cadaba trifoliata leaf extracts against larvae of dengue mosquito Aedes aegypti L. Asian Pac. J. Trop. Biomed. 2012, 2, S1588–S1591. [Google Scholar] [CrossRef]
  67. Punnagai, K.; Chellathai, D.; Latharani, M.; Radhika, R. Evaluation of antibacterial activity of ethanolic extract of Andrographic echioides—An in vitro study. Int. J. Phytopharmacol. 2016, 7, 214–217. [Google Scholar]
  68. Mathivanan, D.; Suseem, S.R. Chemical and biological evaluation of Andrographis echioides leaf extracts collected from the Vellore district in Tamil Nadu, India. Pac. Sci. Rev. A Nat. Sci. Eng. 2016, 18, 138–144. [Google Scholar] [CrossRef]
  69. Gambhire, M.N.; Wnakhede, S.S.; Juvekar, A.R. Antiinflammatory activity of aqueous extract of Barleria cristata leaves. J. Young Pharm. 2009, 1, 220–224. [Google Scholar]
  70. Khare, C.P. Indian Medicinal Plants: An Illustrated Dictionary, 1st ed.; Springer: Berlin, Germany, 2009. [Google Scholar]
  71. Narmadha, R.; Devaki, K. Barleria cristata Linn.: Phytochemical screening and HPTLC analysis. Int. Res. J. Pharm. 2012, 3, 301–307. [Google Scholar]
  72. Abd El-Mawla, A.M.A.; Ahmed, A.S.; Ibraheim, Z.Z.; Ernst, L. Phenylethanoid glycosides from Barleria cristata L. Callus cultures. Bull. Pharm. Sci. Assiut. Univ. 2005, 28, 199–204. [Google Scholar]
  73. Hemalatha, K.; Hareeka, N.; Sunitha, D. Chemical constituents isolated from leaves of Barleria cristata Linn. Int. J. Pharm. Bio. Sci. 2012, 3, 609–615. [Google Scholar]
  74. Chowdhury, N.; Hasan, A.A.; Tareq, F.S.; Ahsan, M.; Zafrul Azam, A.T.M. 4-Hydroxy-trans-cinnamate Derivatives and Triterpene from Barleria cristata. Dhaka Univ. J. Pharm. Sci. 2013, 12, 143–145. [Google Scholar] [CrossRef]
  75. Gambhire, M.; Juvekar, A.; Wankhede, S. Evaluation of anti-inflammatory activity of methanol extract of Barleria cristata leaves by in vivo and in vitro methods. Int. J. Pharmcol. 2008, 7, 1–4. [Google Scholar] [CrossRef]
  76. Amutha, K.; Victor Arokia, D. In vitro antioxidant activity of ethanolic extract of Barleria cristata L. leaves. Res. J. Pharm. Phytochem. 2009, 1, 209–212. [Google Scholar]
  77. Charoenchai, P.; Vajrodava, S.; Somprasong, W.; Mahidol, C.; Ruchirawat, S.; Kittakoop, P. Part 1: Antiplasmodial, cytotoxic, radical scavenging and antioxidant activities of Thai plants in the family Acanthaceae. Planta Med. 2010, 76, 1940–1943. [Google Scholar] [CrossRef]
  78. Sastri, B.N. Wealth of India: Raw Materials; Council of Scientific and Industrial Research: New Delhi, India, 1959; Volume 5, pp. 312–313. [Google Scholar]
  79. Kirtikar, K.R.; Basu, B.D. Indian Medicinal Plants, 2nd ed.; International Book Publisher: Dehradun, India, 1994; Volume 3, pp. 1896–1898. [Google Scholar]
  80. Zhang, H.-J.; Rumschlag-Booms, E.; Guan, Y.-F.; Liu, K.-L.; Wang, D.-Y.; Li, W.-F.; Nguyen, V.H.; Cuong, N.M.; Soejarto, D.D.; Fong, H.H.S.; et al. Anti-HIV diphyllin glycosides from Justicia gendarussa. Phytochemistry 2017, 136, 94–100. [Google Scholar] [CrossRef]
  81. Souza, L.G.S.; Almeida, M.C.S.; Lemos, T.L.G.; Ribeiro, P.R.V.; Canuto, K.M.; Braz-Filho, R.; Cistia, C.N.D.; Sant’Anna, C.M.R.; Barreto, F.S.; Moraes, M.O.D. Brazoides AD new alkaloids from Justicia gendarussa Burm. F. species. J. Braz. Chem. Soc. 2017, 28, 1281–1287. [Google Scholar]
  82. Zhang, H.-J.; Rumschlag-Booms, E.; Guan, Y.-F.; Wang, D.-Y.; Liu, K.-L.; Li, W.-F.; Nguyen, V.H.; Cuong, N.M.; Soejarto, D.D.; Fong, H.H.S.; Rong, L. Potent inhibitor of drug-resistant HIV-1 strains identified from the medicinal plant Justicia gendarussa. J. Nat. Prod. 2017, 80, 1798–1807. [Google Scholar] [CrossRef]
  83. Krishna, K.L.; Mruthunjaya, K.; Patel, J.A. Antioxidant and hepatoprotective potential of stem methanolic extract of Justicia gendarussa Burm. Int. J. Pharmacol. 2010, 6, 72–80. [Google Scholar] [CrossRef]
  84. Bhagya, N.; Chandrashekar, K.R. Evaluation of plant and callus extracts of Justicia gendarussa Burm. F. for phytochemicals and antioxidant activity. Int J. Pharm. Pharm. Sci. 2013, 5, 82–85. [Google Scholar]
  85. Jothimanivannan, C.; Kumar, R.S.; Subramanian, N. Anti-inflammatory and analgestic activities of ethanol extract of aerial parts of Justicia gendarussa Burm. Int. J. Pharmacol. 2010, 6, 278–283. [Google Scholar]
  86. Periyanayagam, K.; Umamaheswari, B.; Suseela, L.; Padmini, M.; Ismail, M. Evaluation of antiangiogenic effect of the leaves of Justicia gendarussa (Burm. f) (Acanthaceae) by Chrio Allontoic Membrane Method. Am. J. Infect. Dis. 2009, 5, 180–182. [Google Scholar] [CrossRef]
  87. Subramanian, N.; Jothimanivannan, C.; Kumar, R.S.; Kameshwaran, S. Evaluation of anti-anxiety activity of Justicia gendarussa Burm. Pharmacologia 2013, 4, 405–407. [Google Scholar]
  88. Paval, J.; Kaitheri, S.K.; Potu, B.K.; Govindan, S.; Kumar, R.S.; Narayanan, S.N.; Moorkoth, S. Anti-arthritic potential of the plant Justicia gendarussa Burm. F. Clinics 2009, 64, 357–360. [Google Scholar] [CrossRef]
  89. Ayob, Z.; Samad, A.A.; Bohari, S.P.M. Cytotoxicity activities in local Justicia gendarussa crude extracts against human cancer cell lines. J. Teknol. Sci. Eng. 2013, 64, 45–52. [Google Scholar] [CrossRef]
  90. Ayob, Z.; Bohari, S.P.M.; Samad, A.A.; Jamil, S. Cytotoxic activities against breast cancer cells of local Justicia gendarussa crude extract. Evid.-Based Complement. Altern. Med. 2014, 2014, 1–12. [Google Scholar] [CrossRef]
  91. Sivasakthi, A.; Vijayalakshmi, M. Antibacterial activities of phytochemical extracts from the leaves of Justicia gendarussa Burm. F. Int. J. Pharm. Biol. Sci. 2014, 5, 433–438. [Google Scholar]
  92. Sharma, K.K.; Saikia, R.; Kotoky, J.; Kalita, J.C.; Devi, R. Antifungal activity of Solanum melogena L., Lawsonia inermis L. and Justicia gendarussa B. against Dermatophytes. Int. J. Pharm. Tech. Res. 2011, 3, 1636–1640. [Google Scholar]
  93. Saha, M.R.; Debnath, P.C.; Rahman, A.; Islam, A.U. Evaluation of in vitro anthelmintic activities of leaf and stem extracts of Justicia gendarussa. Bangladesh J. Pharmacol. 2012, 7, 50–53. [Google Scholar] [CrossRef]
  94. Kumar, K.S.; Vijayan, V.; Bhaskar, S.; Krishnan, K.; Shalini, V.; Helen, A. Anti-inflammatory potential of an ethyl acetate isolated from Justicia gendarussa roots through inhibition of iNOS and COX-2 expression via NF-jB pathway. Cell. Immunol. 2012, 272, 283–289. [Google Scholar] [CrossRef]
  95. Loi, D.T. The Glossary of Vietnamese Medicinal Plants and Items; Hanoi Medicine Publishing House: Hanoi, Vietnam, 2000; pp. 209–210. [Google Scholar]
  96. Gokhani, R.H.; Lahiri, S.K.; Santani, D.D.; Shah, M.B. Evaluation of immunomodulatory activity of Clerodendrum phlomidis and Premna integrifolia root. Int. J. Pharmacol. 2005, 3, 352–356. [Google Scholar]
  97. Basu, N.K.; Dandiya, P.C. Chemical Investigation of Premna integrifolia Linn. J. Amer. Pharm. Assoc. 1947, 36, 389–391. [Google Scholar] [CrossRef]
  98. Vadivu, R.; Suresh, J.A.; Girinath, K.; Kannan, B.P.; Vimala, R.; Kumar, N.M.S. Evaluation of Hepatoprotective and in-vitro cytotoxic activity of leaves of P. serratifolia Linn. J. Sci. Res. 2009, 1, 145–152. [Google Scholar] [CrossRef]
  99. Hang, N.T.B.; Ky, P.T.; Minh, C.V.; Cuong, N.X.; Thao, N.P.; Kiem, P.V. Study on the chemical constituents of Premna integrifolia L. Nat. Prod. Commun. 2008, 3, 1449–1452. [Google Scholar]
  100. Yadav, D.; Tiwari, N.; Gupta, M.M. Diterpenoids from Premna integrifolia. Phytochem. Lett. 2010, 3, 143–147. [Google Scholar] [CrossRef]
  101. Yadav, D.; Gupta, M.M. Isolation and HPTLC analysis of Iridoids in Premna integrifolia, an important ingredient of Ayurvedic drug dashmool. J. Planar Chromatogr. 2013, 26, 260–266. [Google Scholar] [CrossRef]
  102. Yadav, D.; Masood, N.; Luqman, S.; Brindha, P.; Gupta, M.M. Antioxidant furofuran lignans from Premna integrifolia. Ind. Crops Prod. 2013, 41, 397–402. [Google Scholar] [CrossRef]
  103. Rahman, A.; Shanta, Z.S.; Rashid, M.A.; Parvin, T.; Afrin, S.; Khatun, M.K.; Sattar, M.A. In vitro antibacterial properties of essential oil and organic extracts of Premna integrifolia Linn. Arabian J. Chem. 2011, 9, S475–S479. [Google Scholar] [CrossRef]
  104. Majumder, R.; Akter, S.; Naim, Z.; Al-Amin, M.; Alam, M.B. Antioxidant and anti-diabetic activities of the methanolic extract of Premna integrifolia bark. Advan. Biol. Res. 2014, 8, 29–36. [Google Scholar]
  105. Khatun, H.; Majumder, R.; Mamun, A.; Alam, E.K.; Jami, S.I.; Alam, B. Preliminary pharmacological activity of the methanolic extract of Premna integrifolia barks in rats. Avicenna J. Phytomed. 2014, 4, 215–224. [Google Scholar]
  106. Kimura, T.; Kimura, T. Medicinal Plants of Japan in Color; Hoikusha Publishing: Osaka, Japan, 1981; p. 183. [Google Scholar]
  107. Chinese Pharmacopoeia Commission. Pharmacopoeia of the People’s Republic of China; Beijing Chemical Industry Press: Beijing, China, 2010; Volume 1, pp. 340–341. [Google Scholar]
  108. Ono, M.; Sawamura, H.; Ito, Y.; Mizuki, K.; Nohara, T. Diterpenoids from the fruits of Vitex trifolia. Phytochemistry 2000, 55, 873–877. [Google Scholar] [CrossRef]
  109. Ono, M.; Ito, Y.; Nohara, T. Four new halimane-type diterpenes, Vitetrifolins D-G from the fruit of Vitex trifolia. Chem. Pharm. Bull. 2001, 49, 1220–1222. [Google Scholar] [CrossRef]
  110. Li, W.-X.; Cui, C.-B.; Cai, B.; Yao, X.-S. Labdane-type diterpenes as new cell cycle inhibitors and apoptosis inducers from Vitex trifolia L. J. Asian Nat. Prod. Res. 2005, 7, 95–105. [Google Scholar] [CrossRef]
  111. Li, W.-X.; Cui, C.-B.; Cai, B.; Wang, H.-Y.; Yao, X.-S. Flavonoids from Vitex trifolia L. inhibit cell cycle progression at G2/M phase and induce apoptosis in mammalian cancer cells. J. Asian Nat. Prod. Res. 2005, 7, 615–626. [Google Scholar] [CrossRef]
  112. Zheng, C.-J.; Zhu, J.-Y.; Yu, W.; Ma, X.-Q.; Rahman, K.; Qin, L.-P. Labdane-type diterpenoids from the fruits of Vitex trifolia. J. Nat. Prod. 2013, 76, 287–291. [Google Scholar] [CrossRef]
  113. Jangwan, J.S.; Aquino, R.P.; Mencherini, T.; Picerno, P.; Singh, R. Chemical constituents of ethanol extract and free radical scavenging activity of Vitex trifolia Linn. Acta Chim. Pharm. Indica. 2015, 5, 1–7. [Google Scholar]
  114. Tiwari, N.; Luqman, S.; Masood, N.; Gupta, M.M. Validated high performance thin layer chromatographic method for simultaneous quantification of major iridoids in Vitex trifolia and their antioxidant studies. J. Pharm. Biomed. Anal. 2012, 61, 207–214. [Google Scholar] [CrossRef] [PubMed]
  115. Kannathasan, K.; Senthilkumar, A.; Venkatesalu, V. Mosquito larvicidal activity of methyl-p-hydroxybenzoate isolated from the leaves of Vitex trifolia Linn. Acta Trop. 2011, 120, 115–118. [Google Scholar] [CrossRef] [PubMed]
  116. Tandon, S.; Mittal, A.K.; Pant, A.K. Insect growth regulatory activity of Vitex trifolia and Vitex agnus-castus essential oils against Spilosoma obliqua. Fitoterapia 2008, 79, 283–286. [Google Scholar] [CrossRef] [PubMed]
  117. Matsui, M.; Kumar-Roine, S.; Darius, H.T.; Chinain, M.; Laurent, D.; Pauillac, S. Characterisation of the anti-inflammatory potential of Vitex trifolia L. (Labiatae), a multipurpose plant of the Pacific traditional medicine. J. Ethnopharmacol. 2009, 126, 427–433. [Google Scholar] [CrossRef]
  118. Matsui, M.; Conquy, M.A.; Coste, A.; Kumar-Roine, S.; Pipy, B.; Laurent, D.; Pauillac, S. Aqueous extract of Vitex trifolia L. (Labiatae) inhibits LPS-dependent regulation of inflammatory mediators in RAW 264.7 macrophages through inhibition of Nuclear Factor kappa B translocation and expression. J. Ethnopharmacol. 2012, 143, 24–32. [Google Scholar] [CrossRef]
  119. Hernandez, M.M.; Heraso, C.; Villarreal, M.L.; Vargas-Arispuro, I. Biological activities of crude plant extracts from Vitex trifolia L. (Verbenaceae). J. Ethnopharmacol. 1999, 67, 37–44. [Google Scholar] [CrossRef]
  120. Lalchhandama, K. Efficacy and structural effects of Acacia pennata root bark upon the avian parasitic helminth, Raillietina echinobothrida. Pharm. J. 2013, 5, 17–21. [Google Scholar] [CrossRef]
  121. Khare, C.P. Indian Medicinal Plants: An Illustrated Dictionary; Springer: New York, NY, USA, 2008; Volume 8. [Google Scholar]
  122. Rifai, Y.A.; Arai, M.; Kovano, T.; Kowithayakom, T.; Ishibashi, M. Terpenoids and a Flavonoid Glycoside from Acacia pennata Leaves as Hedgehog/GLI-Mediated Transcriptional Inhibitors. J. Nat. Prod. 2010, 73, 995–997. [Google Scholar] [CrossRef]
  123. Dongmo, A.B.; Myyamoto, T.; Yoshikawa, K.; Arihara, S.; Lacaille-Dubois, M.A. Flavonoids from Acacia pennata and their cyclooxygenase (COX-1 and COX-2) inhibitory activities. Planta Med. 2007, 73, 1202–1207. [Google Scholar] [CrossRef]
  124. Kim, A.; Choi, I.; Htwe, K.M.; Chin, Y.-W.; Kim, I.; Yoon, K.D. Flavonoid glycosides from the aerial parts of Acacia pennata in Myanmar. Phytochemistry 2015, 118, 17–22. [Google Scholar] [CrossRef]
  125. Dongmo, A.B.; Nguelefack, T.; Lacaille-Dubois, M.A. Antinociceptive and anti-inflammatory activities of Acacia pennata wild (Mimosaceae). J. Ethnophamacol. 2005, 98, 201–206. [Google Scholar] [CrossRef]
  126. Sowndhararajan, K.; Joseph, J.M.; Manian, S. Antioxidant and free radical scavenging activities of indan Acacias: Acacia leucophloea (Roxb.) Willd., Acacia ferruginea DC., Acacia dealbata Links., and Acacia pennata (L.) Willd. Int. J. Food Prop. 2013, 16, 1717–1729. [Google Scholar] [CrossRef]
  127. Annie, S.; Rajagopal, P.L.; Malini, S. Effect of Cassia auriculata Linn. Root extract on cisplatin and gentamicin-induced renal injury. Phytomedicine 2005, 12, 555–560. [Google Scholar] [CrossRef] [PubMed]
  128. Chauhan, K.N.; Patel, M.B.; Valera, H.R.; Patil, S.D.; Surana, S.J. Hepatoprotective activity of flowers of Cassia curiculata R. Br against paracetamol induced liver injury. J. Nat. Remed. 2009, 9, 85–90. [Google Scholar]
  129. Raj, J.Y.; John Peter, M.P.; Joy, V. Chemical compounds investigation of Cassia auriculata seeds: A potential folklore medicinal plant. Asian J. Plant. Sci. Res. 2012, 2, 187–192. [Google Scholar]
  130. Balakrishna, R.B.; Patel, J.R.; Prabhakaran, V. Cassia auriculata- A phytopharmacological review. J. Adv. Drug Res. 2011, 1, 46–57. [Google Scholar]
  131. Nakamura, S.; Xu, F.; Ninomiya, K.; Nakashima, S.; Oda, Y.; Morikawa, T.; Muraoka, O.; Yoshikawa, M.; Matsuda, H. Chemical Structures and hepatoprotective effects of constituents from Cassia auriculata leaves. Chem. Pharm. Bull. 2014, 62, 1026–1031. [Google Scholar] [CrossRef]
  132. Vijayaraj, P.S.; Muthukumar, K.; Sabarirajan, J.; Nachiappan, V. Evaluation of antihyperlipidemic activity of ethanolic extract of Cassia auriculata flowers. Ind. J. Biochem. Biophys. 2011, 48, 54–58. [Google Scholar]
  133. Gupta, S.; Sharma, S.B.; Bandal, S.K.; Prabhu, K.M. Antihyperglycemic and hypolipidemic activity of aqueous extract of Cassia auriculata L. leaves in experimental diabetes. J. Ethnopharmcol. 2009, 123, 499–503. [Google Scholar] [CrossRef]
  134. Jaydeokar, A.V.; Bandawane, D.D.; Bibave, K.H.; Patil, T.V. Hepatoprotective potential of Cassia auriculata roots on ethanol and natitubercular drug-induced hepatotoxicity in experimental models. Pharm. Biol. 2014, 52, 344–355. [Google Scholar] [CrossRef] [PubMed]
  135. Rajagopal, S.K.; Manickam, P.; Periyasamy, V.; Namasiyayam, N. Activity of Cassia auriculata leaf extract in rats with alcoholic liver injury. J. Nutr. Biochem. 2003, 14, 452–458. [Google Scholar] [CrossRef]
  136. John, C.M.; Sandrasaigaran, P.; Tong, C.K.; Adam, A.; Ramasamy, R. Immunomodulatory activity of polyphenols derived from Cassia auriculata flowers in aged rats. Cell. Immunol. 2011, 271, 474–479. [Google Scholar] [CrossRef] [PubMed]
  137. Sommit, D.; Petsom, A.; Ishikawa, J.; Roengsumran, S. Cytotoxic activity of Natural Labdanes and their semi-synthetic modified derivatives from Croton oblongifolius. Planta Med. 2003, 69, 167–170. [Google Scholar] [CrossRef] [PubMed]
  138. Ngamrojnavanich, N.; Sirimongkon, S.; Roengsumran, S.; Petsom, A.; Kamimura, H. Inhibition of Na+, K+-ATPase activity by (−)-ent-kaar-16-en-19-oic acid and its derivatives. Planta Med. 2003, 69, 555–556. [Google Scholar]
  139. Wijesekera, K. A bioactive diterpene; Nasimalum A from Croton oblongifolius Roxb. Prayog. Ras. 2017, 1, 41–44. [Google Scholar]
  140. Yongsa-ad, W.; Ngamrojanavanich, N.; Mahidol, C.; Ruchirawat, S.; Prawat, H.; Kittakoop, P. Diterpenoids from the roots of Croton oblongifolius. Planta Med. 2007, 73, 1491–1494. [Google Scholar] [CrossRef]
  141. Pudhom, K.; Vilaivan, T.; Ngamrojanayanich, N.; Dechangyipart, S.; Sommit, D.; Petsom, A.; Roengsumran, S. Furanocembranoids from the stem bark of Croton oblongifolius. J. Nat. Prod. 2007, 70, 659–661. [Google Scholar] [CrossRef]
  142. Roengsumran, S.; Petsom, A.; Kuptipyanuwat, N.; Vilaivan, T.; Ngamrojnavanich, N.; Chaichantipyuth, C.; Phuthong, S. Cytotoxic labdane diterpenoids from Croton oblongifolius. Phytochemistry 2001, 56, 103–107. [Google Scholar] [CrossRef]
  143. Roengsumran, S.; Petsom, A.; Sommit, D.; Vilaivan, T. Labdane diterpenoids from Croton oblongifolius. Phytochemistry 1999, 50, 449–453. [Google Scholar] [CrossRef]
  144. Roengsumran, S.; Singtothong, P.; Pudhom, K.; Ngamrochanayanich, N.; Petsom, A.; Chaichantipyuth, C. Neocrotocembranal from Croton oblongifolius. J. Nat. Prod. 1999, 62, 1163–1164. [Google Scholar] [CrossRef] [PubMed]
  145. Roengsumran, S.; Achayindee, S.; Petsom, A.; Pudhom, K.; Singtothong, P.; Surachetapan, C.; Vilaiyan, T. Two new cembranoids from Croton oblongifolius. J. Nat. Prod. 1998, 61, 652–654. [Google Scholar] [CrossRef] [PubMed]
  146. Aiyar, V.N.; Seshadri, T.R. Components of Croton oblongifolius-III constitution of oblongifolic acid. Tetrahedron 1970, 26, 5275–5279. [Google Scholar] [CrossRef]
  147. Rao, P.S.; Sachder, G.P.; Seshadri, T.R.; Singh, H.B. Isolation and constitution of oblongifoliol, a new diterpene of Croton oblongifolius L. Tetrahedron Lett. 1968, 45, 4685–4688. [Google Scholar] [CrossRef]
  148. Adiga, P.G.; Shridhar, N.B.; Sanganal, J.S.; Rao, S.; Shilpa, N.; Aparna, V.; Santhosh, I.M. In vivo anti-tumor efficary of Croton oblongifolius in DMBA induced mammary tumor in rats. Indian J. Anim. Res. 2016, 3301, 1–4. [Google Scholar]
  149. Ahmed, B.; Alam, T.; Varshney, M.; Khan, S.A. Hepatoprotective activity of two plants belonging to the Apiaceae and the Euphorbiaceae family. J. Ethnopharmacol. 2002, 79, 313–316. [Google Scholar] [CrossRef]
  150. Sastri, B.N. The wealth of India: Raw Materials; CSIR: New Delhi, India, 1956; Volume 4, p. 150. [Google Scholar]
  151. Sreejith, P.S.; Praseeja, R.J.; Asha, V.V. A review on the pharmacology and phytochemistry of traditional medicinal plant, Glycosmis pentaphylla (Retz.) Correa. J. Pharm. Res. 2012, 5, 2723–2728. [Google Scholar]
  152. Ito, C.; Kondo, Y.; Rao, K.S.; Tokuda, H.; Nishino, H.; Furukawa, H. Chemical constituents of Glycosmis pentaphylla. Isolation of a novel naphthoquinone and a new acridone alkaloid. Chem. Pharm. Bull. 1999, 47, 1579–1581. [Google Scholar] [CrossRef]
  153. Wang, J.; Di, Y.; Yang, X.; Li, S.; Wang, Y.; Hao, X. Hydroquinone diglycoside acyl esters from the stems of Glycosmis pentaphylla. Phytochemistry 2006, 67, 486–491. [Google Scholar] [CrossRef]
  154. Yang, G.Z.; Wu, Y.; Chen, Y. Alkaloids from the stems of Glycosmis pentaphylla. Helv. Chim. Acta. 2012, 95, 1449–1454. [Google Scholar] [CrossRef]
  155. Chen, Y.; Tian, E.L.; Hu, X.; Li, J.; Yang, G.-Z. Glycopentosides, D-F, three new phenolic glycosides from Glycosmis pentaphylla. Helv. Chim. Acta 2005, 98, 1160–1166. [Google Scholar] [CrossRef]
  156. Chen, Y.; Yang, C.; Wu, Y.; Mo, S.; Wang, S.; Yang, G.; Mei, Z. Glycosmisines A and B: Isolation of two new carbazole-indole-type dimeric alkaloids from Glycosmis pentaphylla and an evaluation of their antiproliferative activities. Org. Biomol. Chem. 2005, 13, 6773–6781. [Google Scholar] [CrossRef] [PubMed]
  157. Sarkar, M.; Chakraborty, D.P. Some minor constituents from Glycosmis pentaphylla. Phytochemistry 1977, 16, 2007–2008. [Google Scholar] [CrossRef]
  158. Mukherjee, S.; Mukherjee, M.; Ganglily, S.N. Glycozolinine, a carbazole derivative from Glycosmis pentaphylla. Phytochemistry 1983, 22, 1064–1065. [Google Scholar] [CrossRef]
  159. Govindachari, T.R.; Pai, B.R.; Subramaniam, P.S. Alkaloids of Glycosmis pentaphylla (Retz.) Correa. Tetrahedron 1966, 22, 3245–3252. [Google Scholar] [CrossRef]
  160. Chowdhury, B.K.; Mustapha, A.; Garba, M.; Bhattacharyya, P. Carbazole and 3-methylcarbazole from Glycosmis pentaphylla. Phytochemistry 1987, 26, 2138–2139. [Google Scholar]
  161. Bhattacharyya, P.; Chakrabarity, P.K.; Chowdhury, B.K. Glycozolidol, and antibacterial carbazole alkaloid from Glycosmis pentaphylla. Phytochemistry 1985, 24, 882–883. [Google Scholar] [CrossRef]
  162. Bhattachryya, P. Glycozolidal, a new carbazole alkaloid from Glycosmis pentaphylla. J. Nat. Prod. 1984, 48, 465–466. [Google Scholar] [CrossRef]
  163. Bhattacharyya, P.; Chowdhury, B.K. Glycolone, a quinolone alkaloid from Glycosmis pentaphylla. Phytochemistry 1985, 24, 634–635. [Google Scholar] [CrossRef]
  164. Zhang, Q.-B.; Ding, G.; Zhang, T.; Si, J.-G.; Song, B.; Wang, M.-H.; Chen, J.-H.; Yu, M.; Gu, Y.-C.; Zou, Z.-M. New furanopyridine alkaloids from the leaves of Glycosmis pentaphylla. Phytochem. Lett. 2006, 18, 51–54. [Google Scholar] [CrossRef]
  165. Sripisut, T.; Ritthiwigrom, T.; Promgool, T.; Yossathera, K.; Deachathai, S.; Phakhodee, W.; Cheenpracha, S.; Laphookhieo, S. Glycopentaphyllone: The first isolation of hydroperoxyquinolone from the fruits of Glycosmis pentaphylla. Phytochem. Lett. 2012, 5, 379–381. [Google Scholar] [CrossRef]
  166. Howlader, A.; Rizwan, F.; Sultana, S.; Rahman, M.R.; Shams-Ud-Doha, K.M.; Mowla, R.; Apu, A.S. Antimicrobial, antioxidant and cytotoxic effects of methanolic extracts of leaves and stems of Glycosmis pentaphylla (Retz.). J. Appl. Pharm. Sci. 2011, 1, 137–140. [Google Scholar]
  167. Quader, M.A.; Nutan, M.T.H.; Rashid, M.A. Antitumor alkaloid from Glycosmis pentaphylla. Fitoterapia 1999, 70, 305–307. [Google Scholar] [CrossRef]
  168. Nayak, S.S.; Jain, R.; Sahoo, A.K. Hepatoprotective activity of Glycosmis pentaphylla against paracetamol-induced hepatotoxicity in Swiss albino mice. Pharm. Biol. 2011, 49, 111–117. [Google Scholar] [CrossRef] [PubMed]
  169. Gupta, N.; Agarwal, M.; Bhatia, V.; Jha, S.K.; Dinesh, J. In vitro antioxidant activity of crude extracts of the plant Glycosmis pentaphylla Correa. Int. J. Pharm. Sci. Rev. Res. 2011, 6, 159–162. [Google Scholar]
Figure 1. Map showing the Myanmar Region.
Figure 1. Map showing the Myanmar Region.
Molecules 24 00293 g001
Figure 2. Structures of selected compounds isolated from Dalbergia culatrata.
Figure 2. Structures of selected compounds isolated from Dalbergia culatrata.
Molecules 24 00293 g002
Figure 3. Structures of selected compounds isolated from Eriosema chinense.
Figure 3. Structures of selected compounds isolated from Eriosema chinense.
Molecules 24 00293 g003
Figure 4. Structures of selected compounds isolated from Erythrina suberosa.
Figure 4. Structures of selected compounds isolated from Erythrina suberosa.
Molecules 24 00293 g004
Figure 5. Structures of selected compounds isolated from Millettia pendula.
Figure 5. Structures of selected compounds isolated from Millettia pendula.
Molecules 24 00293 g005
Figure 6. Structures of selected compounds isolated from Sesbania grandiflora.
Figure 6. Structures of selected compounds isolated from Sesbania grandiflora.
Molecules 24 00293 g006
Figure 7. Structures of selected compounds isolated from Tadehagi triquetrum.
Figure 7. Structures of selected compounds isolated from Tadehagi triquetrum.
Molecules 24 00293 g007
Figure 8. Structures of selected compounds isolated from Andrographis echioides.
Figure 8. Structures of selected compounds isolated from Andrographis echioides.
Molecules 24 00293 g008
Figure 9. Structures of selected compounds isolated from Barleria cristata.
Figure 9. Structures of selected compounds isolated from Barleria cristata.
Molecules 24 00293 g009
Figure 10. Structures of selected compounds isolated from Justicia gendarussa.
Figure 10. Structures of selected compounds isolated from Justicia gendarussa.
Molecules 24 00293 g010
Figure 11. Structures of selected compounds isolated from Premna integrifolia.
Figure 11. Structures of selected compounds isolated from Premna integrifolia.
Molecules 24 00293 g011
Figure 12. Structures of selected compounds isolated from Vitex trifolia.
Figure 12. Structures of selected compounds isolated from Vitex trifolia.
Molecules 24 00293 g012
Figure 13. Structures of selected compounds isolated from Acacia pennata.
Figure 13. Structures of selected compounds isolated from Acacia pennata.
Molecules 24 00293 g013
Figure 14. Structures of selected compounds isolated from Cassia auriculata.
Figure 14. Structures of selected compounds isolated from Cassia auriculata.
Molecules 24 00293 g014
Figure 15. Structures of selected compounds isolated from Croton oblongifolius.
Figure 15. Structures of selected compounds isolated from Croton oblongifolius.
Molecules 24 00293 g015
Figure 16. Structures of selected compounds isolated from Glycosmis pentaphylla.
Figure 16. Structures of selected compounds isolated from Glycosmis pentaphylla.
Molecules 24 00293 g016
Table 1. Scientific name, botanical family and local name of the selected Myanmar medicinal plants.
Table 1. Scientific name, botanical family and local name of the selected Myanmar medicinal plants.
NoScientific NameFamily NameLocal NameRef(s)
1Dalbergia cultrataFabaceaeYin-daik[4]
2Eriosema chinenseFabaceaePeik-san-gale[4]
3Erythrina suberosaFabaceaeKa-thit[4]
4Millettia pendulaFabaceaeThin-win[4]
5Sesbania grandifloraFabaceaePaukpan-phyu[4]
6Tadehagi triquetrumFabaceaeLauk-thay or shwe-gu-than-hlet[4]
7Andrographis echioidesAcanthaceaeSega-gyi-hmwe-tu[4]
8Barleria cristataAcanthaceaeLeik-tha-ywe-pya[4]
9Justicia gendarussaAcanthaceaePha-wa-net[4]
10Premna integrifoliaVerbenaceaeTaungtan-gyi[4]
11Vitex trifoliaVerbenaceaeKyanung-ban[4]
12Acacia pennataMimosaceaeSuyit or Suboke-gyi[4]
13Cassia auriculataCaesalpiniaceaePeik-thingat[4]
14Croton oblongifoliusEuphorbiaceaeThetyin-gyi[4]
15Glycomis pentaphyllaRutaceaeTaw-shauk[4]
Table 2. Biological/pharmacological activities of compounds isolated from selected medicinal plants.
Table 2. Biological/pharmacological activities of compounds isolated from selected medicinal plants.
SourceCompoundBiological/Pharmacological ActivitiesReference(s)
D. culrata (Stem bark)Dalberatin A (6)Cancer chemopreventive activity with IC50 of 212 (mol ratio/32 pmol TPA)[10]
D. culrata (Stem bark)Dalberatin B (7)Cancer chemopreventive activity with IC50 of 303 (mol ratio/32 pmol TPA)[10]
E. chinense (Roots)Khonklonginol A (8)Cytotoxicity against KB (IC50 3.1 μg/mL), NCI-H187 (IC50 3.0 μg/mL), Antimycobacterial activity against Mycobacterium tuberculosis H37Ra (MIC 25 μg/mL), Antimicrobial activity against Bacillus cereus (MIC 150 μg/mL), Staphylococcus agalactiae (MIC 2.3 μg/mL) and S. pyrogenes (MIC 2.3 μg/mL), Antioxidant activity (IC50 7.919 mM)[11,12]
E. chinense (Roots)Khonklonginol B (9)Cytotoxicity against KB (IC50 3.8 μg/mL), NCI-H187 (IC50 4.3 μg/mL)[11]
E. chinense (Roots)Lupinifolinol (16)Cytotoxicity against KB (IC50 1.73 μg/mL), NCI-H187 (IC50 3.5 μg/mL), Antimicrobial activity against Candia albicans (IC50 75 μg/mL), Bacillus cereus (IC50 4.7 μg/mL), Listeria monocytogenes (IC50 9.4 μg/mL), Staphylococcus aureus (IC50 75 μg/mL), S. aureusRASA (IC50 9.4 μg/mL), S. agalactiae (IC50 4.7 μg/mL), S. epidermidis (IC50 9.4 μg/mL), S. pyrogenes (IC50 2.3 μg/mL), Antioxidant activity (IC50 1.768 mM)[11,12]
E. chinense (Roots)Dehydrolupinifolinol (17)Antimycobacterial activity against Mycobacterium tuberculosis H37Ra (MIC 12.5 μg/mL)[11]
E. chinense (Roots)Flemichin D (18)Antimycobacterial activity against Mycobacterium tuberculosis H37Ra (MIC 12.5 μg/mL), Antimicrobial activity against with the values of (IC50 ˃ 150 μg/mL) each for Candida albicans, Escherichia coli, Klebsiella pneumoniae and Pseudomonas aeruginosa and (IC50 4.7 μg/mL) for each Bacillus cereus, Enterococcus faecalis, Listeria monocytogenes, Staphylococcus aureus, S. aureus RASA, S. agalactiae, S. epidermidis and S. pyrogenes, Antioxidant activity (IC50 0.538 mM)[11,12]
E. chinense (Roots)Eriosemaone A (19)Antimycobacterail activity against Mycobacterium tuberculosis H37Ra (MIC 12.5 μg/mL)[11]
E. chinense (Roots)Lupinifolin (20)Antimycobacterial activity against Mycobacterium tuberculosis H37Ra (MIC 12.5 μg/mL)[11]
E. chinense (Roots)3-epi-lupinifolinol (21)Antioxidant activity (IC50 0.681 mM)[12]
E. chinense (Roots)2′-dihydroxy lupinifolinol (23)Antimicrobial activity against Candia albicans (IC50 37.5 μg/mL), Escherichia coli (IC50 75 μg/mL), Klebsiella pneumoniae (IC50 75 μg/mL), and Pseudomonas aeruginosa (IC50 75 μg/mL), Bacillus cereus (IC50 2.3 μg/mL), Enterococcus faecalis (IC50 9.4 μg/mL), Listeria monocytogenes (IC50 9.4 μg/mL), Staphylococcus aureus (IC50 4.7 μg/mL), S. aureusRASA (IC50 4.7 μg/mL), S. agalactiae (IC50 4.7 μg/mL), S. epidermidis (IC50 37.5 μg/mL), S. pyrogenes (IC50 2.3 μg/mL), Antioxidant activity (IC50 0.252 mM)[12]
E. chinense (Roots)3,5,2′,4′-Tetrahydroxy-6″,6″ dimethylpyrano (2″,3″:7,6)-8-(3‴,3‴-dimethylallyl) flavone (24)Antimicrobial activity against Candia albicans (IC50 75 μg/mL), Escherichia coli (IC50 ˃ 150 μg/mL), Klebsiella pneumoniae (IC50 150 μg/mL), and Pseudomonas aeruginosa (IC50 150 μg/mL), Bacillus cereus (IC50 9.4 μg/mL), Enterococcus faecalis (IC50 18.8 μg/mL), Listeria monocytogenes (IC50 18.8 μg/mL), Staphylococcus aureus (IC50 9.4 μg/mL), S. aureusRASA (IC50 9.4 μg/mL), S. agalactiae (IC50 9.4 μg/mL), S. epidermidis (IC50 9.4 μg/mL), S. pyrogenes (IC50 9.4 μg/mL), Antioxidant activity (IC50 0.035 mM)[12]
E. chinense (Roots)Tectorigenin (28)Antimicrobial activity against with the values of (IC50 > 150 μg/mL) for each Candida albicans, Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, Bacillus cereus, Enterococcus faecalis, Listeria monocytogenes, Staphylococcus aureus, S. aureusRASA, S. agalactiae, S. epidermidis and S. pyrogenes, Antioxidant activity (IC50 3.666 mM)[12]
E. chinense (Roots)Genistein (29)Antimicrobial activity against Candida albicans (IC50 75 μg/mL) and (IC50 150 μg/mL) for each Klebsiella pneumoniae, Pseudomonas aeruginosa, Bacillus cereus, Staphylococcus aureus, S. agalactiae and S. pyrogenes[12]
E. chinense (Roots)Kaempferol (30)Antimicrobial activity against with the values of (IC50 > 150 μg/mL) each for Candida albicans, Klebsiella pneumoniae, Pseudomonas aeruginosa, Bacillus cereus, Staphylococcus aureus, S. agalactiae and S. pyrogenes, Antioxidant activity (IC50 0.028 mM)[12]
E. chinense (Roots)Kaempferol-7-O-β-d-glucopyranoside (31)Antimicrobial activity against with the values of (IC50 > 150 μg/mL) for each Candida albicans, Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, Bacillus cereus, Listeria monocytogenes, Staphylococcus agalactiae and S. epidermidis, Antioxidant activity (IC50 0.651 mM)[12]
E. chinense (Roots)Astragalin (32)Antimicrobial activity against with the values of (IC50 150 μg/mL) for each Candida albicans, Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, Bacillus cereus, Enterococcus faecalis, Staphylococcus aureus, S. agalactiae, S. epidermidis and S. pyrogenes, Antioxidant activity (IC50 0.681 mM)[12]
E. suberosa (Stem bark)4′-methoxy licoflavanone (36)The cytotoxic effects on apoptosis in human leukemia HL-60 cells and their potency to induce cancer cell death[19]
E. suberosa (Stem bark)Alpinumisoflavone (37)The cytotoxic effects on apoptosis in human leukemia HL-60 cells and their potency to induce cancer cell death[19]
E. suberosa (Stem bark)Erysodine (40)The anxiolytic effects in the elevated plus-maze and the light-dark transition model[15]
E. suberosa (Flowers)Erysotrine (41)The anxiolytic effects in the elevated plus-maze and the light-dark transition model[15]
M. pendula (Timber)Millettilone A (49)Leishmanicidal activity (IC50 9.3 μg/mL)[25]
M. pendula (Timber)3R-Claussequinone (50)Leishmanicidal activity (IC50 1.2 μg/mL)[25]
M. pendula (Timber)Pendulone (51)Leishmanicidal activity (IC50 0.07 μg/mL)[25]
M. pendula (Timber)Secundiflorol I (53)Leishmanicidal activity (IC50 86 μg/mL)[25]
M. pendula (Timber)3,8-Dihydroxy-9-methoxy pterocarpan (54)Leishmanicidal activity (IC50 2.9 μg/mL)[25]
M. pendula (Timber)3,10-Dihydroxy-7,9-dimethoxypterocarpan (55)Leishmanicidal activity (IC50 77 μg/mL)[25]
S. grandiflora (Roots)Isovestitol (57)Antituberculosis activity against M. tuberculosis H37Rv (MIC 50 μg/mL)[27]
S. grandiflora (Roots)Medicarpin (58)Antituberculosis activity against M. tuberculosis H37Rv (MIC 50 μg/mL)[27]
S. grandiflora (Roots)Sativan (59)Antituberculosis activity against M. tuberculosis H37Rv (MIC 50 μg/mL)[27]
S. grandiflora (Roots)Betulinic acid (60)Antituberculosis activity against M. tuberculosis H37Rv (MIC 100 μg/mL)[27]
T. treiquetrum (whole plant)Tadehaginosin (69)Hypoglycemic activity in vitro by HepG2 cells[45]
T. treiquetrum (whole plant)3,4-Dihydro-4-(4′-hydroxyphenyl)-5,7-dihydroxycoumarin (70)Hypoglycemic activity in vitro by HepG2 cells[45]
T. treiquetrum (Aerial part)Tadehaginosides C–J (7380)Antidiabetic activity[46]
T. treiquetrum (Aerial part)Tadehaginoside (81)Antidiabetic activity[46]
A. echioides (whole plant)Dihydroechioidinin (82)Anti-inflammatory activity with the IC50 of 37.6 ± 1.2 μM[58]
A. echioides (whole plant)5,7,8-Trimethoxyflavone (98)Anti-inflammatory activity with the IC50 of 39.1 ± 1.3 μM[58]
J. gendarussa (Stem and Bark)Justiprocumin B (111)Anti-HIV activity against a broad spectrum of HIV strains with IC50 values in the range of 15–21 nM (AZT, IC50 77–95 nM), nevirapine resistant isolate HIV-1N119 with an IC50 value of 495 nM and AZT resistant isolate HIV-11617-1 with (IC50 185 nM)[80]
J. gendarussa (Stem and Bark)Patentiflorin A (116)Anti-HIV activity against a broad spectrum of HIV strains with IC50 values in the range of 24-37 nM (AZT, IC50 77–95 nM), drug-resistant HIV-1 isolate of both the nucleotide analogue (AZT) and (nevirapine)[82]
P. integrifolia (Stem bark)10-O-trans-p-Methoxycinnamoyl catalpol (119)Antioxidant activity with the IC50 value of 0.37 μM/mL in DPPH free radical scavenging assay[101]
P. integrifolia (Stem bark)4″-Hydroxy-E-globularinin (125)Antioxidant activity with the IC50 value of 0.29 μM/mL in DPPH free radical scavenging assay[101]
P. integrifolia (Stem bark)Premnosidic acid (126)Antioxidant activity[101]
P. integrifolia (Stem bark)Premnadimer (127)Antioxidant activity[101]
P. integrifolia (Stem bark)4β-Hydroxyasarinin-1-O-β-glucopyranoside (128)Antioxidant activity[101]
V. trifolia (Aerial part)Agnuside (152)Antioxidant activity (IC50 9.81 μg) in DPPH and (IC50 12.90 μg) NO radical scavenging assays[114]
V. trifolia (Aerial part)Negundoside (153)Antioxidant activity (IC50 9.96 μg) in DPPH and (IC50 16.25 μg) NO radical scavenging assays[114]
V. trifolia (Aerial part)6-p-Hydroxybenzoyl mussaenosidic acid (154)Antioxidant activity (IC50 10.31 μg) in DPPH and (IC50 13.51 μg) and NO radical scavenging assays[114]
V. trifolia (Leaves)Methyl-p-hydroxybenzoate (155)Mosquito larvicidal activity against LC50 values of methyl-p-hydroxybenzoate were 5.77 ppm against Culex quinquefasciatus and 4.74 ppm against Aedes aegypti[115]
A. pennata (Leaves)Taepeenin D (156)Hedgehog/GLi-mediated transcriptional activity with IC50 value of 1.6 Μm, Cytotoxic against pancreatic (PANC1) cells (IC50 3.2 μM) and prostate (DU145) cells (IC50 3.4 μM)[122]
A. pennata (Leaves)(+)-Drim-8-ene (157)Hedgehog/GLi-mediated transcriptional activity with IC50 value of 13.5 μM, Cytotoxic against pancratic (PANC1) cells (IC50 15.1 μM) and prostate (DU145) cells (IC50 23.2 μM)[122]
A. pennata (Leaves)Quercetin 3-O-β-d-glucopyranosyl-4-O-β-d-glucopyranoside (158)Hedgehog/GLi-mediated transcriptional activity with IC50 value of 10.5 μM, Cytotoxic against pancreatic (PANC1) cells (IC50 26.6 μM) and prostate (DU145) cells (IC50 30.0 μM)[122]
C.auriculata (Leaves)Pseudosemiglabrin (187)Hepatoprotective effects [inhibition % 37.0 ± 3.7 (p ˂ 0.01)] at 30 μM[135]
C. auriculata (Leaves)(2S)-7,4-Dihydroxy flavan (4β→8)-catechin (188)Hepatoprotective effects [inhibition % 33.5 ± 2.9 (p < 0.01)] at 30 μM[135]
C. auriculata (Leaves)(2S)-7,4-Dihydroxy flavan (4β→8)-gallocatechin (189)Hepatoprotective effects [inhibition % 28.2 ± 4.7 (p < 0.01)] at 30 μM[135]
C. auriculata (Leaves)(−)-Epigallocatechin (192)Hepatoprotective effects [inhibition % 24.4 ± 3.1 (p < 0.01)] at 30 μM[135]
C. oblongifolius (Leaves)Nasimalun A (193)Cytotoxicity toward MOLT-3 cell line with IC50 26.44 μg/mL, Antibacterial activity MIC value of 50 μg/mL for Bacilus cereus and 100 μg/mL for Staphylococcus aureus and Staphylococcus epidermidis[139]
C. oblongifolius (Stem bark)Furanocembranoid 1 (201)Cytotoxic effects against human tumor cell lines BT474 (IC50 7.8 μg/mL), CHAGO (IC50 7.0 μg/mL), Hep-G2 (IC50 5.6 μg/mL), KATO-3 (IC50 5.9 μg/mL) and SW-620s (IC50 6.3 μg/mL) by MTT colorimetric method[142]
C. oblongifolius (Stem bark)Furanocembranoid 3 (203)Cytotoxic effects against human tumor cell lines BT474 (IC50 9.6 μg/mL), CHAGO (IC50 7.1 μg/mL), Hep-G2 (IC50 5.7 μg/mL), KATO-3 (IC50 8.2 μg/mL) and SW-620s (IC50 5.6 μg/mL) by MTT colorimetric method[142]
C. oblongifolius (Stem bark)Furanocembranoid 4 (204)Cytotoxic effects against human tumor cell lines BT474 (IC50 9.6 μg/mL), CHAGO (IC50 9.3 μg/mL), Hep-G2 (IC50 6.1 μg/mL), KATO-3 (IC50 8.1 μg/mL) and SW-620s (IC50 6.0 μg/mL) by MTT colorimetric method[142]
C. oblongifolius (Stem bark)(−)-ent-kuar-16-en-19-oic acid (205)Inhibition of Na+, K+-ATPase activity with an IC50 of 2.2 × 10−5 M[138]
C. oblongifolius (Stem bark)Croblongifolin (206)Cytotoxic effects against human tumor cell lines including HEP-G2 (IC50 0.35 μM), BT474 (IC50 0.12 μM), SW-620 (IC50 0.47 μM) CHAGO (IC50 0.24 μM) and KATO-3 (IC50 0.35 μM)[143]
C. oblongifolius (Stem bark)Neocrotocembranal (215)Cytotoxicity against P-388 cell culture in vitro (IC50 6.48 μg/mL)[145]
G. pentaphylla (Stem)Biscarbalexine A (227)Cytotoxicity against human cancer cell lines A549 (IC50 56.06 μM), HepG-2 (IC50 60.06 μM) and Huh-7 (IC50 73.16 μM)[156]
G. pentaphylla (Stem)Glycosmisine A (231)Cytotoxicity against human cancer cell lines A549 (IC50 43.68 μM), HepG-2 (IC50 50.30 μM) and Huh-7 (IC50 30.60 μM)[156]
G. pentaphylla (Stem)Glycosmisine B (232)Cytotoxicity against human cancer cell lines A549 (IC50 57.10 μM), HepG-2 (IC50 62.89 μM) and Huh-7 (IC50 62.87 μM)[156]

Share and Cite

MDPI and ACS Style

Aye, M.M.; Aung, H.T.; Sein, M.M.; Armijos, C. A Review on the Phytochemistry, Medicinal Properties and Pharmacological Activities of 15 Selected Myanmar Medicinal Plants. Molecules 2019, 24, 293. https://doi.org/10.3390/molecules24020293

AMA Style

Aye MM, Aung HT, Sein MM, Armijos C. A Review on the Phytochemistry, Medicinal Properties and Pharmacological Activities of 15 Selected Myanmar Medicinal Plants. Molecules. 2019; 24(2):293. https://doi.org/10.3390/molecules24020293

Chicago/Turabian Style

Aye, Mya Mu, Hnin Thanda Aung, Myint Myint Sein, and Chabaco Armijos. 2019. "A Review on the Phytochemistry, Medicinal Properties and Pharmacological Activities of 15 Selected Myanmar Medicinal Plants" Molecules 24, no. 2: 293. https://doi.org/10.3390/molecules24020293

Article Metrics

Back to TopTop