You are on page 1of 20

View Online / Journal Homepage / Table of Contents for this issue

REVIEW www.rsc.org/npr | Natural Product Reports

Recent advances in anti-HIV natural products


Inder Pal Singh* and Hardik S. Bodiwala
Received 12th August 2010
DOI: 10.1039/c0np00025f

Covering: 2004 to April 2010

This review covers the recent advances in anti-HIV natural products of various origins and chemical
classes. The literature of the last seven years has been thoroughly reviewed, with around 160
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F

compounds discussed and 141 references cited. Some molecules in an advanced stage of development,
such as calanolide A and bevirimat, are also mentioned.

1 Introduction due to AIDS and preventing new infections. The number of new
Downloaded by University of Sussex on 06 August 2012

2 Natural products from plants HIV infections declined from 3.0 million in 2001 to 2.7 million in
2.1 Alkaloids 2008,1 and the efforts of the World Bank, WHO and G8 decla-
2.1.1 Newly discovered anti-HIV alkaloids ration for universal access to treatment can be regarded as the
2.1.2 Known alkaloids discovered to be anti-HIV agents key factors for this positive result.2 However, overall, 2.0 million
2.2 Phenolics people died in 2008 due to HIV infection compared with an
2.2.1 Newly discovered anti-HIV phenolics estimated 1.7 million in 2001,1 a fact that can be attributed to the
2.2.2 Known phenolics discovered to be anti-HIV agents higher number of infections in the early part of this decade
2.3 Terpenes coupled with lesser availability of newer and efficacious anti-
2.3.1 Newly discovered anti-HIV terpenes retroviral drugs during those years in the sub-Saharan region.
2.3.2 Known terpenes discovered to be anti-HIV agents Currently, the treatment available for the disease is known as
2.4 Proteins and polysaccharides HAART (Highly Active Anti-Retroviral Therapy). However,
2.5 Miscellaneous compounds due to complexity and social stigma associated with HIV in
3 Natural products of microbial origin developing countries, the long-term management of HIV
3.1 Newly discovered anti-HIV agents of microbial patients is difficult. The potent drugs may fail at a later stage
origin because of chronic adverse effects and the emergence and
3.2 Known microbial metabolites discovered to be anti- transmission of drug-resistant variants. This underlines the need
HIV agents for continuous efforts in the discovery of new antiretroviral/anti-
4 Natural products of marine origin HIV agents.3
5 Advanced-stage molecules – where are they now? Many of the reviews published earlier on this topic are mainly
6 Conclusions and future perspectives on a specific class of compounds, or natural products active
7 Acknowledgement against specific targets, such as integrase. For example, there are
8 References reviews with limited coverage such as those on coumarins,4 tri-
terpenes,5 flavanoids,6 integrase inhibitors at Merck,7 and
natural products with anti-HIV activity from marine sponges.8
The most recent review by Cos et al. covers natural anti-HIV
1 Introduction compounds reported during 1997–2007,9 although many of the
references are either prior to 2005 or synthetic modifications or
Human Immunodeficiency Virus (HIV) / AIDS remains a major
further mechanistic studies on previously isolated anti-HIV
health problem worldwide that has already caused an estimated
compounds. The present review provides extensive coverage of
25 million deaths and has generated profound demographic
all natural products obtained over the last seven years (2004–
changes in the most heavily affected countries in the last 27 years.
2010) from diverse sources that exhibit anti-HIV activity towards
Globally, there were an estimated 33 million people living with
different biological targets.
HIV in 2008. Young people between 15 to 24 years of age
The HIV replication cycle is summarized in Fig. 1. The first
account for an estimated 45% of the new HIV infections
step in the life-cycle involves virus fusion to the host cell through
worldwide. A 6-fold increase in financing for HIV programmes
viral glycoprotein gp120 and CD4 receptors. Then, gp120
in low- and middle-income countries is beginning to give results,
interacts with the chemokine co-receptor (either CCR5 or
with many countries making major progress in lowering deaths
CXCR4). This is followed by release of the viral RNA into the
host cell cytoplasm. By action of the reverse transcriptase
Department of Natural Products, National Institute of Pharmaceutical enzyme, viral RNA is transcribed into double-stranded viral
Education and Research (NIPER), Sector-67, S.A.S. Nagar, Punjab,
160062, India. E-mail: ipsingh@niper.ac.in; ipsingh67@yahoo.com; Fax: DNA that migrates into the host cell nucleus. This viral DNA is
+91 172-2214692; Tel: +91 172-2292144 then incorporated into the host cell chromosome by the action of

This journal is ª The Royal Society of Chemistry 2010 Nat. Prod. Rep., 2010, 27, 1781–1800 | 1781
View Online
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F
Downloaded by University of Sussex on 06 August 2012

Fig. 2 Newly discovered anti-HIV alkaloids.

Fig. 1 The HIV life-cycle showing the different targets. alkaloids were evaluated and discovered to be anti-HIV agents.
The structures of anti-HIV alkaloids are shown in Fig. 2 and 3.
integrase. Following this, various viral proteins and viral
genomic material are produced after transcription and trans-
2.1.1 Newly discovered anti-HIV alkaloids. A diprenylated
lation by the host cell. Finally, two copies of viral RNA and
indole alkaloid, (E)-3-(3-hydroxymethyl-2-butenyl)-7-(3-methyl-
other proteins are incorporated into the immature virus, which is
2-butenyl)-1H-indole (1), was isolated from the twigs and leaves
then released and converted into a mature and functionally active of Glycosmis montana native to China. The alkaloid 1 exhibited
virus by the Gag polyprotein. potent anti-HIV activity with an IC50 value of 1.17 mg/mL and an
SI of 11.68. It was suggested that these diprenylated indole
alkaloids might be candidates for further study as potential anti-
2 Natural products from plants HIV agents.10 A sesquiterpene-pyridine alkaloid, tripfordine A
(2), was isolated from an ethanolic extract of the roots of
2.1 Alkaloids
poisonous liana Tripterygium wilfordii. The highly toxic roots are
Alkaloids of various structural subclasses have been reported to known as ‘lei gong teng’ in Chinese folklore and used as an
possess anti-HIV activity. During the coverage period, few new insecticide. Compound 2 possesses an unusual structure con-
alkaloids were reported as anti-HIV agents, but many known taining a sesquiterpene skeleton joined to a pyridine ring by

Inder Pal Singh obtained his Hardik S. Bodiwala was born


Ph.D. from Punjab Agricultural in 1982 in Ahmedabad,
University, Ludhiana, India. In Gujarat, India. After
1994, he moved to Shizuoka completing a B.Pharm. in
University, Japan, on a Mon- 2003, he obtained his
busho Fellowship and obtained M.S.(Pharm.) from NIPER in
his second Ph.D. under the 2005. During this period, he
guidance of Prof. Hideo Etoh. worked on the isolation and
He worked as a post-doctoral characterization of antipara-
associate (1998–2000) in Prof. sitic compounds. In 2005 and
Gerwick’s laboratory at Oregon 2006 he worked in the R&D
State University, USA. Later, unit of Cadila Pharmaceuticals
he joined Institute of Chemical Ltd, Ahmedabad, on method
Inder Pal Singh Research, Kyoto University, Hardik S: Bodiwala development and analysis of
Japan, on a JSPS fellowship for biological samples. He is
a two-year period. He is currently pursuing a Ph.D.
currently an Associate Professor at NIPER. His research interests under the supervision of Inder Pal Singh on the discovery of
include isolation of bioactive molecules from natural sources, natural product based anti-HIV agents.
biomimetic synthesis of bioactive natural products, and standard-
ization of herbal/Ayurvedic formulations.

1782 | Nat. Prod. Rep., 2010, 27, 1781–1800 This journal is ª The Royal Society of Chemistry 2010
View Online
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F
Downloaded by University of Sussex on 06 August 2012

Fig. 3 Known alkaloids discovered to be anti-HIV agents.

a macrocycle. Compound 2, along with the other sesquiterpene- suggested that the binding site of anibamine had some features in
pyridine alkaloids, were screened against HIV-1. Tripfordine A common with the other CCR5 antagonists, therefore suggesting
showed moderate anti-HIV activity, with an EC50 of 13.4 mg/mL that 3 may bind in similar binding sites and/or modes.13
and TI of 1.9, while the other alkaloids exhibited more potent A new canthin-4-one type alkaloid, drymaritin (4), isolated from
activity, with EC50 values of 2.54 to <0.1 mg/mL. The SAR Drymaria diandra (also known as Drymaria cordata subsp. diandra)
suggested that the C-5 acetyl group may be essential for the exhibited anti-HIV activity in H9 lymphocytes with an EC50 value
activity, as compounds with a 5-acetyl group were more active of 0.699 mg/mL and a TI of 20.6. The canthin-6-one type of alka-
than those with 5-hydroxy, 5-benzyloxy and 5-furanoyl groups. loids have been already reported to have anti-HIV activity.14
The position of the carboxypropyl or carboxybutyl side chain
(C-20 or C-40 ) on the nicotinic moiety did not affect the activity 2.1.2 Known alkaloids discovered to be anti-HIV agents. The
(EC50 <0.10 mg/mL). Further, the compounds with a tertiary iboga-type indole alkaloid 18-methoxycoronaridine (18-MC) (5)
hydroxy group in the propionic (C-80 ) or butyric (C-90 ) acid side was semisynthesized from coronaridine, and inhibits retroviral
chain showed anti-HIV activity, while those without an OH were activity with EC50 of 22.5 and 23 mM for R5 and X4 isolates in
inactive or less active. However, one alkaloid hyponine B, which PBMC. Similarly, 5 inhibited HIV-1 replication in macrophages
lacks an OH on C-80 , was also highly active; hence, it was sug- with EC50 values of 12.8 and 9.5 mM and TI of 25.6 and 34.5 after
gested that other structural parameters may also play an 14 and 21 days of infection, respectively. It moderately inhibited
important role in the activity.11 HIV-1 RT with an IC50 of 69.4 mM.15 The major metabolite of 5
Anibamine (3), a novel pyridine quaternary alkaloid, was in human liver microsomes was 18-hydroxycoronaridine
isolated from the CH3OH extract of the stems of an Aniba species (18-HC). The metabolism of 18-MC to 18-HC was catalyzed by
as a TFA salt. Anibamine competed for the binding of 125I-gp120 the polymorphic enzyme CYP2C19, and studies of this suggested
to human CCR5 with an IC50 of 1 mM.12 Molecular docking that 18-MC may be a selective probe substrate of CYP2C19, one
studies of anibamine in two different CCR5 homology models that is perhaps more selective and specific than currently used
(bovine rhodopsin and human b2-adrenergic receptors) probes (omeprazole and mephenytoin).16 Two carbazole

This journal is ª The Royal Society of Chemistry 2010 Nat. Prod. Rep., 2010, 27, 1781–1800 | 1783
View Online
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F
Downloaded by University of Sussex on 06 August 2012

Fig. 4 Newly discovered anti-HIV phenolics.

alkaloids, glybomine B (6) and glycoborinine (7), were isolated 4.47 mg/mL, respectively.10 Other carbazole alkaloids, O-meth-
from the twigs and leaves of Glycosmis montana. Earlier these ylmukonal (8), 3-formyl-2,7-dimethoxycarbazole (9) and claus-
alkaloids were reported from G. arborea.17,18 Both the alkaloids zoline J (10), were isolated from the rhizomes and roots of
exhibited weak to moderate in vitro inhibitory activity against Clausena excavata. Compounds 8–10 showed anti-HIV-1 activity
HIV replication in C8166 cells. Compounds 6 and 7 demon- in a syncytial assay, with EC50 values of 12.0, 29.1 and 34.2 mM
strated anti-HIV activity with an IC50 of 9.73 mg/mL and and TI of 56.7, 8.0 and 1.6, respectively.19

1784 | Nat. Prod. Rep., 2010, 27, 1781–1800 This journal is ª The Royal Society of Chemistry 2010
View Online
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F
Downloaded by University of Sussex on 06 August 2012

Fig. 5 Known phenolics discovered to be anti-HIV agents.

This journal is ª The Royal Society of Chemistry 2010 Nat. Prod. Rep., 2010, 27, 1781–1800 | 1785
View Online
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F
Downloaded by University of Sussex on 06 August 2012

Fig. 6 Known phenolics discovered to be anti-HIV agents (continued).


1786 | Nat. Prod. Rep., 2010, 27, 1781–1800 This journal is ª The Royal Society of Chemistry 2010
View Online

The alkaloids decarine (11), g-fagarine (12) and (+)-temba- showed in vitro inhibitory effects against HIV replication in H9
mide (13) were the most potent anti-HIV compounds of 67 lymphocytes, with an EC50 of 11.3 mM.31 Longipedunin A (28)
compounds isolated from the root bark of Zanthoxylum ailan- was isolated from the roots and stems of Kadsura long-
thoides. Compounds 11–13 showed EC50 values of <0.1 mg/mL ipedunculata, and showed an IC50 of 50 mg/mL against HIV-1
and TI values of >226, >231, and >215, respectively. Fourteen PR. The higher activity of 28 compared to other dibenzocy-
other compounds from the same plant showed significant activity clooctadiene lignans indicated that a trans-cinnamic acid ester
against HIV replication in H9 lymphocyte cells.20 group might be an important functional group for the anti-HIV
Two benzylisoquinoline alkaloids, (+)-1-(R)-coclaurine (14) PR activity.32 Tiegusanin G (29) was one of 14 new lignans iso-
and ()-1-(S)-norcoclaurine (15), isolated from the leaves of lated from a 70% aqueous acetone extract of the aerial parts of
Nelumbo nucifera, a perennial aquatic crop grown throughout Schisandra propinqua var. sinensis, popularly known as ‘tie-gu-
Asia, showed anti-HIV activity with EC50 values of 0.8 and san’ in China. Compound 29 showed anti-HIV-1 activity in the
<0.8 mg/mL and TI values of >125 and >25, respectively. C8166 cell line, with an EC50 value of 7.9 mM and TI of > 25.33
Compound 15 is a precursor in the biosynthesis of well-known New flavonoid glycosides 6-(g,g-dimethylallyl)dihydro-
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F

anti-HIV alkaloids such as berberine and aporphine, as well as kaempferol-7-O-b-D-glucoside (30), 6-(g,g-dimethylallyl)dihydro-
other biologically active benzylisoquinoline and benzophenan- quercetin-7-O-b-D-glucoside (31), 6-(3-hydroxy-3-methylbutyl)
thridine alkaloids.21 Norcoclaurine synthase is the first enzyme in taxifolin-7-O-b-D-glucoside (32) and 6-(3-hydroxy-3-methylbutyl)-
the biosynthetic pathway to benzylisoquinoline alkaloids in quercetin-7-O-b-D-glucoside (33) were isolated from the anti-
several plants.22 Some other bisbenzylisoquinoline alkaloids, HIV-active methanolic extract of Ochna integerrima. These
Downloaded by University of Sussex on 06 August 2012

liensinine (16), negferine (17) and isoliensinine (18), were evalu- compounds showed anti-HIV-1 activity in the syncytium assay
ated against HIV to establish the SAR. Compounds 16–18 with an EC50 of 29.1, 14.0, 102.4 and 45.8 mg/mL, respectively. In
showed anti-HIV activity with EC50 values of <0.8 mg/mL and TI the same study, vitexin (containing a sugar unit on ring A) was
values of >9.9, >8.6 and >6.5, respectively, while the aporphine found to be inactive. Earlier, Meragelman et al.34 had reported
alkaloid nuciferine (19) showed an EC50 of 0.8 mg/mL and TI of that 6-isoprenylated flavonoids with two hydroxyls at C-5 and
36.23 Norruffscine (20) and ()-8-oxotetrahydropalmatine (21) C-7 were inactive while McKee et al.35 reported that compounds
were isolated from Pericampylus glaucus, a climbing shrub widely with 30 -OH were active against HIV. This suggested that the
distributed in the southwest of China. Compounds 20 and 21 isoprenyl and sugar unit on ring A and 30 -OH (which are present
showed EC50 values of 10.9 and 14.1 mM and SI values of 45.7 in the above compounds) are required for anti-HIV activity.36
and 18.8 respectively against HIV-1 in C8166 cells.24 Three phenolic compounds, 2,3-dihydroxy-4-methoxy-6,6,9-tri-
Cycleanine (22) a bisbenzylisoquinoline alkaloid isolated from methyl-6H-dibenzo[b,d]pyran (34), 8-methoxy-2-methyl-2-(4-
the root bark of Epinetrum villosum, was active against HIV-2 methyl-3-pentenyl)-2H-1-benzopyran-6-ol (35) and 4-methoxy-
(EC50 1.83 mg/mL); however, it was 10 times less active against 3-(3-ethyl-2-butenyl)benzoic acid (36) isolated from Wigandia
HIV-1.25 Cycleanine is also reported to show potent antibacte- urens showed activity in the CCR5 assay with IC50 values of 33,
rial, antifungal, antiplasmodial, and cytotoxic activities. Tablets 46 and 26 mM respectively. Compound 36 was previously
containing 2 mg cycleanine caused inhibition of TPA-promoted synthesized as an intermediate.37 Arzanol (37) is a phloroglucinol
tumours in rat ear, as well as superoxide generation by poly- a-pyrone derivative isolated form the acetone extract of Heli-
morphonuclear leukocytes. Cycleanine-containing hair tonic was chrysum italicum ssp. microphyllum, one of the best-known
shown to control dandruff and prevent alopecia.26 Amary- medicinal plants in the Mediterranean area. The structural
llidaceae alkaloids lycorine (23) and homolycorine (24) isolated elucidation of arzanol led to a revision of the structure of
from the bulbs of Leucojum vernum possess anti-HIV-1 activity in homoarenol. Arzanol inhibited TNFa-induced HIV-LTR
MT4 cells with ID50 values of 0.4 and 7.3 mg/mL, respectively. In transactivation in a T-cell line with an IC50 of 5 mM, but failed to
addition, 23 was active in a mechanism-based anti-HIV-RT inhibit luciferase activity in HeLa Tet-On-Luc assay.38
assay, with an ID50 of 0.4 mg/mL.27 Lycorine is also reported to The dimeric lactone ardimerin digallate (38) isolated from the
show antimalarial activity towards Plasmodium falciparum.28 A whole plants of Ardisia japonica, showed inhibitory activity
small molecule, indole-3-carboxylic acid (25), was among 44 against both HIV-1 and HIV-2 RNase H in vitro with IC50 values
known compounds isolated from rhizomes of Begonia nantoen- of 1.5 and 1.1 mM, respectively. It did not inhibit human or
sis, and was also active against HIV replication in H9 lympho- E. coli RNase H at 200 mM, demonstrating selectivity of 38
cyte cells, with an EC50 of 2.41 mg/mL and a TI of 6.79.29 towards inhibition of the retroviral enzyme. A similar compound
lacking a galloyl moiety, ardimerin, did not show any inhibition
of RNase H, suggesting that the galloyl group is important for
2.2 Phenolics
anti-HIV activity.39
Various phenolics, such as calanolide A, DCK and DCQA, are Three quinic acid derivatives, methyl-5-O-caffeoyl-3-O-sina-
currently in clinical trials.30 The structures of anti-HIV phenolics poylquinate (39), methyl-5-O-caffeoyl-4-O-sinapoylquinate (41)
are shown in Figs. 4–6. and methyl-3,5-di-O-caffeoyl-4-O-(3-hydroxy-3-methyl)glutar-
oylquinate (43), were isolated by bioactivity-guided fraction-
2.2.1 Newly discovered anti-HIV phenolics. An inseparable ation of the ethyl acetate extract of Gardenia jasminoides
2 : 1 mixture of new dibenzocyclooctadiene lignans rubrisandrins (Gardeniae Fructus). Compounds 39, 41 and 43 showed HIV-1
A (26 + 27) was isolated from the fruits of Schisandra rubriflora, IN inhibitory activity with an IC50 of 20 mg/mL. Ethyl quinate
indigenous to southern China. The two regioisomers were derivatives 40 and 42 showed an IC50 of 45 mg/mL in the same
distinguished by 1H, 13C and 2D-NMR data. The mixture assay.40

This journal is ª The Royal Society of Chemistry 2010 Nat. Prod. Rep., 2010, 27, 1781–1800 | 1787
View Online

2.2.2 Known phenolics discovered to be anti-HIV agents. diarrhoea, abnormal uterine bleeding, edema, anemia, albu-
Sixteen known compounds, including dibenzocyclooctadiene- minuria in chronic nephritis, and diabetes. Compound 70
type lignans, were isolated from the stems of Kadsura heteroclita, inhibited cytopathic effects with an EC50 of 15.7 mM and TI of
a plant native to southern China. Lignans kadsurin (44), heter- 28.5.49
oclitin F (45) and acetoxy oxokadsurane (46) were active as anti- Biflavonoids ochnaflavone 700 -O-methyl ether and 200 ,300 -
HIV agents. Compounds interiorin (47) and interiorin B (48) dihydroochnaflavone 700 -O-methyl ether, isolated from Ochna
exhibited anti-HIV activity with EC50 values of 1.6 and integerrima, were found to be active in the syncytium assay with
1.4 mg/mL, and TI values of 52.9 and 65.9, respectively. EC50 values of 2.0 and 0.9 mg/ml, respectively. In a mechanism-
Comparison of 47 and 48 with other dibenzocyclooctadiene based assay, these compounds inhibited HIV-1 RT with IC50
lignans having similar skeletons indicated that the aa0 bb0 -dien- values of 2.0 and 2.4 mg/mL, respectively.36
one might be an important functional group for anti-HIV Orobol (71) and wedelolactone (72) isolated from Eclipta
activity.41 prostrata showed activity against HIV-1 IN with IC50 values of
Dibenzocyclooctadiene lignans gomisin J (49), ()-gomisin 8.1 and 4.0 mM, respectively; but they did not inhibit HIV-1 PR.50
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F

M1 (50), (+)-gomisin M2 (51) and schisanhenol (52) were isolated Wedelolactone possesses a wide range of biological activities,
from the fruits of Schisandra rubriflora. ()-Gomisin M1 (50) including its use as an antidote for snake venom and hep-
exhibited the most potent anti-HIV activity, with EC50 and TI atoprotective effects. It is also known to inhibit the IKK
values of <0.65 mM and >68, respectively. It was found that complex.51 The cyclohexenyl chalcone hydroxypanduratin A (73)
compounds with aromatic hydroxyl groups were more active has been obtained from the methanolic extract of rhizomes of
Downloaded by University of Sussex on 06 August 2012

than those lacking aromatic hydroxyls, suggesting the impor- Boesenbergia pandurata, a perennial herb of the Zingiberaceae
tance of the aromatic hydroxyl groups for the anti-HIV activity family. This plant is locally known in Thailand as ‘kra-chai’, and
of dibenzocyclooctadiene lignans. The position of the hydroxyl has been used as self-medication by AIDS patients in that
was also important for enhancing the anti-HIV activity, as 50 country. Compound 73 possesses anti-HIV-1 protease activity,
(with a 30 -OH), was more potent than 51 (with a 3-OH).31 Four with an IC50 value of 5.6 mM. SAR of cyclohexenyl chalcones for
known lignans, gomisin J (49), schizandrin (53), schisantherin A HIV-1 PR suggested that compounds with a hydroxyl group at
(54) and schisantherin D (55), were isolated from the leaves and C-4 were more active than those with a methoxy group, preny-
stems of Schisandra sphenanthera. The anti-HIV-1 activity of 49 lated compounds were more active than non-prenylated
and 53–55 was evaluated by the inhibition assay for the cyto- compounds, hydroxylation at the 4000 -position reduced activity,
pathic effects of HIV-1IIIB. Compounds 49 and 53–55 showed while introduction of a double bond at C10 and C60 increased the
anti-HIV-1 activity, with EC50 values of 17.2, 15.5, 19.0 and activity against HIV-PR.52
20.5 mg/mL, respectively.42 Gomisin J is also known to inhibit Punicalin (74) and 2-O-galloylpunicalin (75), isolated from the
L-type calcium channels and stimulate glucose uptake into aqueous extract of Terminalia triflora leaves showed inhibitory
peripheral tissue.43,44 Anti-inflammatory activity in murine activity on HIV-1 RT, with an IC50 of 0.14 mM and 0.11 mM.
macrophages has also been reported.45 Binakadsurin A (56) was Both the tannins were previously isolated from the genus
isolated from 70% aqueous acetone extract of stems of Kadsura Terminalia.53 Punicalins and punicalagins, as well as other
angustifolia, and was isolated along with 11 new lignans. tannins, are well-known antioxidants and anticancer agents,54,55
Compound 56 showed anti HIV-1 activity in the C8166 cell line, and have also shown antimicrobial and antimalarial activities.56
with an EC50 of 3.86 mM, a CC50 of 227.16 mM and an SI of Clausenidin (76), a pyranocoumarin, was isolated from the
58.92.46 rhizomes and roots of Clausena excavata and it displayed anti-
Flavonoids quercetin (57), quercitrin (58) and myricetin 3-O- HIV-1 activity in a syncytial assay with an EC50 of 5.3 mM and
b-D-galactopyranoside (59) were isolated from the EtOAc extract a TI of 7.0. Earlier, 76 was reported to be inactive in an HIV-RT
of leaves of Alnus firma. The compounds showed inhibition assay, so it might be acting on a different target in the HIV life-
against HIV-1 RT with an IC50 of 60 mM, whereas they were cycle.19
inactive against HIV-1 PR.47 5,7-Dihydroxychromone (60), and 4-Phenylcoumarins were isolated from lipophilic dichloro-
()-catechin (61), isolated from Begonia nantoensis, endemic to methane extract of leaves of Marila pluricostata collected from
the mountains of central Taiwan, demonstrated activity against Panama.57 Mesuol (77) affected neither the reverse transcription
HIV replication in H9 lymphocyte cells with EC50 values of 18.7 nor integration steps of the viral cycle, however the antiviral
and 14.3 mg/mL, and TI values of 1.3 and 1.8, respectively.29 activity was additive when combined with AZT. A mechanistic
Eight prenylisoflavonoids were isolated from the DCM extract study revealed that mesuol inhibited TNFa-induced HIV-1–
of Erythrina senegalensis, a Cameroonian medicinal plant. LTR transcriptional activity by targeting the NF-kB pathway.
8-Prenylluteone (62), auriculatin (63), erysenegalensein O (64), This hinted at the potential of the NF-kB transcription factor as
erysenegalensein D (65), erysenegalensein N (66), derrone (67), one of the targets for anti-HIV compounds.58 In addition,
alpinumisoflavone (68) and 6,8-diprenylgenistein (69) showed compounds 78, 79 and 80 were found to be active in the NF-kB
HIV-1 PR inhibitory activity with IC50 values of 4.0, 3.5, 5.0, 2.5, and HeLa-Tat-Luc tests at 25 mM concentration. These
4.5, 18.2, 30.1 and 0.5 mM, respectively. The two prenyl groups at compounds were active against recombinant HIV with IC50
the 6- and 8-positions, along with a hydroxy at the 40 -position, values of 6.9, 31.9 and 0.5 mM, respectively.59
were found to be important for the activity.48 Calycosin-7-O-b-D- The caged xanthones morellic acid (81), gambogic acid (82),
glucopyranoside (70) has been isolated from the roots of desoxygambogenin (83), forbesione (84) and dihydroisomorellin
Astragalus membranaceus var. mongholicus. This plant has been (85) were isolated from the resin and fruits of Garcinia hanburyi.
used as traditional Chinese medicine to cure various diseases like Compounds 83 and 85 showed anti-HIV-1 activity in

1788 | Nat. Prod. Rep., 2010, 27, 1781–1800 This journal is ª The Royal Society of Chemistry 2010
View Online

a syncytium assay with EC50 values of 3.0 and 1.2 mg/mL and TI polyprenylated benzophenones structurally similar to clusianone
values of 1.7 and 4.7, respectively. Compounds 81, 82, 84 and 85 are reported to possess antioxidant activities.64
exhibited anti-HIV-1 RT activity, with IC50 values of 11.0, 15.0, Long-chain phenols have been isolated from Ginkgo biloba
62.1 and 42.3 mg/mL.60 Two xanthones, 1,3,7-trihydroxy-6- sarcotestas. Bilobal C15:1 (93) and C13:0 (94) were active against
methoxy-4,5-di-(3-methylbut-2-enyl)xanthone (86) and euxan- HIV-1 PR, with IC50 values of 2.6 and 5.8 mM, respectively.
thone (87), were isolated form the MeOH extract of the aerial Alkylsalicylic acid C15:1 (95), C17:1 (96) and C15:0 (97) were found
part of Cratoxylum arborescens. Compounds 86 and 87 displayed to be active against HIV-1 PR, with IC50 values of 24.9, 17.8 and
anti-HIV-1 RT activity with an IC50 of 84.9 and 50.1 mg/mL, 10.2 mM. Compounds 95 and 96 were also found to be active
respectively.61 against HIV-RT, with IC50 values of 33.7 and 58.5 mM, respec-
Polyisoprenylated benzophenones were isolated from the tively. Bilobals possessing two hydroxy groups at the meta
fruits of Clusia torresii, a tree endemic to Costa Rica. Clusianone position were found to be more active against HIV-1 PR than
(88), 7-epi-clusianone (89), 18,19-dihydroxyclusianone (90), alkylsalisylic acids (one hydroxyl and one carboxylic acid) or
propolone A (91) and nemorosone (92) have shown anti-HIV carbanols (only one hydroxyl group). Alkylsalicylic acids with
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F

activity with EC50 values of 0.02, 2.0, 7.1, 0.32 and 0.8 mM, and one carboxylic acid group exhibited moderate activity against
SI values of 5.0, 10.0, 2.2, 15.6 and 6.2, respectively. Compounds RNase from HIV-1 RT.65
88 and 89 remain in constant equilibrium between their keto and
enol tautomers. The SAR suggested that presence of a keto–enol
equilibrium was not essential for anti-HIV activity, and that the 2.3 Terpenes
Downloaded by University of Sussex on 06 August 2012

C-7 configuration might play an important role in the activity. A


study of the mechanism of action revealed that benzophenone Lee et al. have recently reviewed plant-derived anti-HIV tri-
derivatives of type B (88–90) inhibited the gp120-sCD4 interac- terpenes.66 We have included here those terpenes that were not
tion, indicating that they might be interfering with the viral reported in this earlier review. The structures of anti-HIV
attachment to the cellular receptor CD4, and thus preventing terpenes are shown in Figs. 7 and 8.
infection. Compounds 91 and 92 had no effect on the gp120-
sCD4 interaction.62 Clusianone, first discovered in 1976, is also
known to have cancer-chemopreventive properties.63 Other

Fig. 7 Newly discovered anti-HIV terpenes. Fig. 8 Known terpenes discovered to be anti-HIV agents.

This journal is ª The Royal Society of Chemistry 2010 Nat. Prod. Rep., 2010, 27, 1781–1800 | 1789
View Online

2.3.1 Newly discovered anti-HIV terpenes. Three new tri- and showed activity against HIVNL4-3 at 5 mg/mL. Forskolin, the
terpenoids, (20R)-20,25-epoxydammaran-2-en-6a,12b-diol (98), major constituent of this plant, is reported to show various
(20R)-20,25-epoxy-3-methyl-28-nordammaran-2-en-6a,12b-diol biological activities, including cardiotonic, antihypertensive, and
(99) and isodehydroprotopanaxatriol (100) were isolated from an adenylcyclase activator.74 Sesquiterpenes oxyphyllendiol B (111)
acidic hydrolysate of Panax ginseng. These compounds showed and 1,2,3,4-tetrahydro-2,5-dimethyl-8-(1-methylethyl)-1,2-
inhibitory activity against HIV-1 PR, with IC50 values of 10.5, naphthalenediol (112) were isolated from Litesea verticillata.
10.3 and 12.3 mM, respectively.67 Two new lupanes, 2a-acetoxy- Compunds 111 and 112 were found to inhibit HIV-1 replication
3b-hydroxy-19b-H-lup-20(29)-en-28-oic acid (2-acetoxy- in the GFP-based cell line HOG.R5, with IC50 values of 54.6 and
alphitolic acid) (101) and 2a-hydroxy-3b-acetoxy-19b-H-lup- 91.0 mM, respectively.71
20(29)-en-28-oic acid (3-acetoxyalphitolic acid) (102) were iso-
lated from the EtOAc extract of resins of Garcinia hanburyi
2.4 Proteins and polysaccharides
collected from Thailand. Lupanes 101 and 102 showed anti-HIV-
1 activity in the syncytium assay with EC50 values of 5.6 and 6.0 Aqueous and 80% ethanolic extracts of 20 Thai medicinal plants
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F

mg/mL and SI values of 2.8 and 3.3, respectively. They also dis- used to treat AIDS were tested for their HIV-1 RT inhibitory
played anti-HIV-1 RT inhibitory activity, with IC50 values of activity. The aqueous extracts of Ipomoea carnea subsp. fistulosa
116.9 and 16.3 mg/mL, respectively.68 (aerial parts), Vitex glabrata (branches), Vitex trifolia (aerial
Schilancidilactone A (103), a novel tetranortriterpenoid parts), Vitex negundo (aerial parts), Canna indica (rhizomes), and
possessing an unprecedented C26 skeleton (and thus representing Justicia gendarussa (aerial parts) showed HIV-1 RT inhibition in
Downloaded by University of Sussex on 06 August 2012

a new class of Schisandra nortriterpenoids), was isolated from the a radiometric assay at 200 mg/mL. Bioactivity-guided fraction-
70% aqueous acetone extract of stems of Schisandra lancifolia. ation of an aqueous extract of Canna indica rhizomes (IC50
Compound 103 showed anti-HIV-1 activity, with an EC50 of 8.51 22.6 mg/mL) yielded two proteins, Cip31 (31 kDa) and Cip14
mg/mL and an SI >23.5.69 (14 kDa). Both the proteins were found to be active against HIV-
Highly functionalized ingol diterpene 8-methoxyingol-7,12- 1 RT, with IC50 values of 17.4 and 19.2 mg/mL, respectively.75
diacetate-3-phenylacetate (104) was isolated from Euphorbia Juncin, an 18.9 kDa protein, was isolated from seeds of Brassica
officinarum latex collected from plants on the North Atlantic juncea var. integrifolia, and showed activity against HIV-1 RT
coast of Morocco at Agadir. Compound 104 caused cell-cycle with an IC50 of 4.5 mM.76 A 38 kDa protein was isolated from
arrest and HIV-1-LTR promoter activation through a common fresh Capparis spinosa melon seeds. It exhibited partial similarity
signalling pathway in a concentration-dependent manner, and in an N-terminal amino acid sequence with imidazole glycerol
was proposed as a new lead for the development of therapies phosphate synthase, and inhibited HIV-1 RT with an IC50 of
against HIV-1 latency.70 0.23 mM. The mechanism of inhibition may be protein–protein
The sesquiterpene isolitseane B (105) was isolated from the interaction; HIV-1 PR is reported to inhibit the homologous
chloroform extract of the leaves and twigs of Litesea verticillata retroviral RT in a similar fashion.77 Griffithsin, a novel anti-HIV
collected from Vietnam. Compound 105 was found to inhibit protein with 121 amino acids and a molecular weight of
HIV-1 replication, with an IC50 of 38.1 mM and an SI of 3; the 6a 13 kDa, was isolated from an aqueous extract of the red alga
isomer was found to be inactive.71 Griffithsia sp. collected from the waters of New Zealand. Grif-
fithsin displayed antiviral activity against laboratory strains and
2.3.2 Known terpenes discovered to be anti-HIV agents. Two primary isolates of T- and M-tropic HIV-1, with EC50 values
cucurbitacins, 25-O-acetyl-23, 24-dihydrocucurbitacin F and ranging from 0.043 to 0.63 nM. Griffithsin also stopped cell
dihydrocucurbitacin F, named hemslecins A (106) and B (107) fusion and transmission of HIV-1 infection at similar concen-
respectively, were isolated from tubers of a Chinese medicinal trations.78
plant, Hemsleya jinfushanensis. These compounds were earlier Several cyclotides, macrocyclic plant peptides containing three
isolated from many plants of this genus, which are used to cure disulfide bonds, have been reported as anti-HIV agents. Various
inflammatory diseases such as enteritis, diarrhoea and bronchitis. cyclotides have been isolated from Viola yedoensis, a Chinese
Both 106 and 107 inhibited syncytia formation with EC50 values herb. The isolated cyclotides kalata B1, varv E, and cycloviolacin
of 3.09 and 2.53 mg/mL, and p24 antigen production with EC50 Y1, Y4 and Y5 were active in an XTT-based anti-HIV assay in
values of 3.97 and 18.90 mg/mL, respectively. They also inhibited the range 0.04–1.2 mM. The most active, cycloviolacin Y5, was
fusion between HIV-1-infected H9 cells and C8166 cells with also the most hydrophobic peptide of all the isolated compounds.
EC50 values of 1.8 and 11.9 mg/mL. Although these compounds Therefore, a positive correlation between the hydrophobicity of
showed slight interaction with HIV-1 IN, they did not inhibit cyclotides and anti-HIV activity was inferred.79
HIV-1 RT, PR, or NCp7 zinc ejection. It was concluded that 106 Polysaccharide fractions TK-V2, 3 and 4, containing spirulan-
and 107 block or compete for the binding site of HIV-1, or like substances, were isolated from Arthrospira platensis. TK-V2
modulate the molecular expression which is necessary for HIV-1 contained a high quantity of carbohydrates while TK-V3 was
binding to reduce entry of the virus. The acetyl group was found composed of 41% carbohydrates and 57% proteins. TK-V4 was
to enhance the anti-HIV-1 activities of this type of compounds.72 the soluble part of precipitated TK-V3, containing 45% carbo-
3,4-Seco-(24Z)-cycloart-4(28),24-diene-3,26-dioic acid 26-methyl hydrates. All fractions showed an inhibitory effect against HIV-
ester (108) was isolated from Illicium verum, and inhibited HIV- 1NL4-3 at 90 mg/mL. TK-V3 and 4 showed IC50 values of 3.2 and
1IIIB with an EC50 of 5.1 mM.73 2.1 mg/mL, respectively.80
Labdane diterpenes forskolin (109) and 1-deoxyforskolin (110) A dimeric 64 kDa meliobiose-binding lectin was isolated from
were isolated from the chloroform extract of Coleus forskohlii the seeds of Bauhinia variegata. This lectin inhibited HIV-1

1790 | Nat. Prod. Rep., 2010, 27, 1781–1800 This journal is ª The Royal Society of Chemistry 2010
View Online

reverse transcriptase with an IC50 of 1.02 mM.81 The mannose-


specific lectins from Galanthus nivalis (GNA) and a Hippeastrum
hybrid (HHA) occur as tetramers of four 12.5 kDa peptides.
Further, they were not glycosylated, and were present as
a mixture of isomers. Both lectins exhibited antiviral activity
against HIV-1 and HIV-2 in CEM cell cultures, with EC50 values
between 0.12 and 1.2 mg/mL for GNA and between 0.18 and
0.70 mg/mL for HHA, depending on the nature of the virus
strain.82 A 60 kDa lectin from seeds of the autumn purple bean
(Phaseolus vulgaris) was isolated and the sequence of its first 20
N-terminal amino acids was identified as ANEIYFSFQRF-
NETNLILQR. The lectin inhibited HIV-1 RT with an IC50 of
1.8 mM.83 BanLec, a jacalin-related lectin isolated from the fruit
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F

of the banana Musa acuminata, exists as a dimer with a molecular


mass of 30 kDa. BanLec showed anti-HIV activity, with IC50
values in the low nanomolar to picomolar range in different
assays with various isolates. BanLec inhibited HIV-1 infection by
binding to the glycosylated viral envelope and blocking cellular
Downloaded by University of Sussex on 06 August 2012

entry.84

2.5 Miscellaneous compounds


A new benzofuranone lactone, 5-hydroxy-3-(propan-2-ylidene)-
7-(3,7,11,15-tetramethylhexa-deca-2,6,10,11-tetraenyl)-2(3H)-
benzofuran, named rhuscholide A (113), was isolated from the
ethanolic extract of stems of Rhus chinensis. It demonstrated
anti-HIV-1 activity, with an EC50 of 1.62 mM and a TI of 42.4. A Fig. 9 Miscellaneous anti-HIV compounds.
similar compound lacking the 3-(propan-2-ylidene) group was
found to be less active and more toxic (EC50 3.7 and TI 3.2) in
3.1 Newly discovered anti-HIV agents of microbial origin
a cell-based assay.85 These compounds did not inhibit HIV-RT
and PR in an enzyme-based assay. Furthermore, in a co-culti- Cytochalasins are a group of fungal secondary metabolites with
vation assay, these compounds did not inhibit cell–cell fusion. various biological activities. Alachalasin A (121) was isolated
Thus, it was proposed that this type of compound might target from the cultures of an isolate of Stachybotrys charatum collected
the late steps of the HIV life-cycle.86 Butanolides 3-epi-litsenolide from a Tibetan glacier. Compound 121 showed inhibitory effect
D2 (114) and litseabutenolide (115) were isolated from the leaves on HIV-1LAI replication in C8166 cells, with an EC50 of
and twigs of Litesea verticillata, and were found to inhibit HIV-1 8.01 mM.87 Cytochalasins show antimicrobial activity, inhibition
replication in the GFP-based cell line HOG.R5 with IC50 values of glucose transport, regulate plant growth and are known to
of 9.9 and 40.3 mM.71 ()-Illicinone A (116) was isolated from inhibit angiogenesis.88 Pestalofones A (122), B (123) and E (124)
Illicium verum, and inhibited HIV-1IIIB with an EC50 of are new cyclohexanone derivatives isolated from cultures of the
16.0 mM.73 plant endophytic fungus Pestalotiopsis fici and displayed inhib-
5-Hydroxymethyl-(2,20 :50 ,200 )terthienyl tiglate (117), itory effects on HIV-1 replication in C8166 cells, with EC50
5-hydroxymethyl-(2,20 :50 ,200 )terthienyl agelate (118), 5-hydroxy- values of 90.4, 64.0 and 93.7 mM, respectively. All three
methyl-(2,20 :50 ,200 )terthienyl acetate (119) and ecliptal (120) were compounds showed CC50 values of >200 mM.89
isolated from the whole plant extract of Eclipta prostrata, Three compounds, 10-methoxydihydrofuscin (125), fuscinarin
a perennial herb which grows widely throughout tropical areas of (126) and fuscin (127), were isolated from the soil fungus
Asia. Compounds 117, 119 and 120 were active against HIV-1 Oidiodendron griseum. Compound 127 was isolated previously
PR with IC50 values of 58.3, 93.7 and 83.3 mM. Compound 117 from various species of genus Oidiodendron. Compounds 125–
which is in the cis form, exhibited activity, whereas 118, which is 127 showed competitive inhibition of the binding of macrophage
in the trans form, was inactive (IC50 >100 mM) against HIV-1 inflammatory protein (MIP)-1a to human CCR5 with IC50
PR. This suggests that the geometry of double bond plays an values of 154, 80 and 21 mM, respectively, in the scintillation
important role in activity.50 The structures are shown in Fig. 9. proximity binding assay. The higher activity of 127 compared to
125 and 126 was ascribed to the presence of multiple reactive sites
in the isochromene-5,9-dione unit.90
Azaphilones phomoeuphorbin A (128) and phomoeuphorbin
3 Natural products of microbial origin
C (129), isolated from the cultures of Phomopsis euphorbiae, an
Singh et al. have reviewed HIV integrase inhibitors of mircobial endophytic fungus isolated from Trewia nudiflora, were tested
origin.7 The structures of microorganism-derived anti-HIV for in vitro inhibitory effects against HIV replication in C8166
natural products are shown in Figs. 10 and 11. cells. Compounds 128 and 129 showed anti-HIV activities with

This journal is ª The Royal Society of Chemistry 2010 Nat. Prod. Rep., 2010, 27, 1781–1800 | 1791
View Online
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F
Downloaded by University of Sussex on 06 August 2012

Fig. 10 Newly discovered anti-HIV agents of microbial origin.

EC50 values of 79 mg/mL and 71 mg/mL and cytotoxicity isolated from a fruiting body collected in Puerto Rico yielded
against C8166 cells at CC50 >200 mg/mL. However, the ther- 19,20-epoxycytochalasin Q (134). Both compounds exhibited
apeutic index of >2.5 was not favourable for their future binding of 125I-gp120 to human CCR5 with an IC50 of 40 and
development.91 Helotialins A–C (130–132) were isolated as new 60 mM, respectively.12 Two CCR-5 inhibitors, Sch 210971 (135)
azaphilones from the cultures of an ascomycete of the order and Sch 210972 (136) were isolated from the fungus Chaetomium
Helotiales. Compounds 130 and 131 displayed inhibitory globosum. Compounds 135 and 136 had IC50 values of 1.2 mM
effects on HIV-1 replication in C8166 cells with EC50 values of and 79 nM respectively in the CCR-5 receptor binding assay.93
8.01 and 27.9 mM, respectively. Compound 132 was inactive. It Variecolin (137) and variecolol (138), isolated from a 1 : 1
was observed that the C-13–C-14 olefin might be important CH2Cl2–CH3OH extract of the fungus Emericella aurantio-
for anti-HIV activity.92 brunneai, had IC50 values of 9 and 32 mM respectively for binding
A culture of the fungus Mollisia sp., which was isolated from to human CCR5 in a scintillation proximity assay. Differences in
the dead bark of Tsuga canadensis collected on Hamburg IC50 values suggested that the C-4 ketone and the C-20 aldehyde
Mountain, NJ, USA, by Merck Research Laboratories yielded might be important for activity against the human CCR5
ophiobolin C (133), while another fungal culture of Xylaria sp. receptor.94

1792 | Nat. Prod. Rep., 2010, 27, 1781–1800 This journal is ª The Royal Society of Chemistry 2010
View Online
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F
Downloaded by University of Sussex on 06 August 2012

Fig. 11 Known microorganism-derived natural products discovered to be anti-HIV agents.

3.2 Known microbial metabolites discovered to be anti-HIV fuscoatra. SCH 642305 (142) was produced from fungus Septo-
agents fusidium sp. collected from Puerto Rico, having earlier been
reported from Penicillium verrucosum. Compounds 140–142
Hinnuliquinone (139) a C2-symmetric bis-indolyl quinone, was
inhibited HIV-1 Tat transactivation with IC50 values of 0.027, 5.0
isolated from the fungus on the leaves of Quercus coccifera
and 1.0 mM. Compound 142 was more toxic to the Jurkat cell line
collected in Spain. Hinnuliquinone, originally isolated from
than 140 and 141 (IC50 4.10 mM vs. $100 mM). Similar molecules
Nodulisporium hinnuleum, inhibited both the wild-type and
lacking a keto group in the macrocyclic ring were found to be
a clinically resistant (A44) strain of HIV-1 PR, with IC50 values
inactive. The sesquiterpenoids (+)-sporogen AO1 (143) and
of 2.5 and 1.8 mM, respectively. Simultaneously, some related
(+)-petasol (144) were isolated from EtCOMe extract of Pen-
compounds having an isomeric quinone moiety or benzoquinone
icillum sp. from a soil sample. Compound 143 was first isolated
were screened for anti-HIV PR activity, and were found to be
from Hansfordia pulvinata and 144 was isolated from a whole-
inactive. It was therefore assumed that the activity was not
plant extract of Petasites fragrans. These compounds inhibited
simply because of the quinone moiety, but some other structural
HIV-1 Tat transactivation, with IC50 values of 15.8 and 46.2 mM,
features of the molecule might be responsible for the activity.95
respectively.
Merck laboratories have identified several natural products
Echinomycin (145; quinomycin A) is a quinoxiline bicyclic
active against HIV-1 by using a HIV-1 Tat protein dependent in
octadepsipeptide produced by a Streptomyces sp. collected from
vitro cell-based assay. The identified compounds include the
Japan. It was reported to be a HIV-1 RT inhibitor with an IC50
macrocyclic lactones monorden (140), monocillin IV (141) and
of 700 mM.96 Sulfonium salts of echinomycin showed potent
SCH 642305 (142), the sesquiterpenoids sporogen AO1 (143) and
antitumour activity, while similar octadepsipeptides show anti-
petasol (144), the bicyclic octadepsipeptides echinomycin (145)
bacterial activity.97 UK-63598 (146) is a quinoline bicyclic octa-
and UK-63598 (146), and a cyclic heptapeptide, ternatin (147).96
depsipeptide isolated from the EtCOMe extract of
Monorden (140), also known as radicicol, and monocillin IV
Dactylosporangium sp., earlier reported from Streptomyces
(141) were produced by Penicillium sp. Compound 140 was
braegenesis. Compound 146 inhibited Tat transactivation with
originally isolated from Monosporium bonorden and subse-
an IC50 of 0.2 nM and was non-cytotoxic to Jurkat cells up to
quently it was reported from several fungal species. Monocillin
0.05 mM, whereas 145 was slightly less active, with an IC50 of
IV (141) was reported from Monocillum nordinii and Humicola
1.5 nM and a CC50 of 0.025 mM. Ternatin (147) was isolated

This journal is ª The Royal Society of Chemistry 2010 Nat. Prod. Rep., 2010, 27, 1781–1800 | 1793
View Online

from EtCOMe extract of the fermented broth of a fungus found to inhibit the cytopathic effect of HIV-1, with an EC50 of
belonging to the Xylariaceae family found on the leaves of 28 nM in a cell-based XTT assay. Direct cytotoxicity of 155
Quercus ilex, and inhibited Tat transactivation with an IC50 of against the host cells was observed, with a TC50 of 260 nM.106
0.3 mM.96 Some other cyclic depsipeptides, mirabamides A–D, were iso-
lated from marine sponge material collected from Nama Island,
Micronesia. Mirabamides A, C and D (156–158) inhibited HIV-1
4 Natural products of marine origin
in a neutralization assay in two different viral strains (HXB2 and
A new water-soluble flavonoid glycoside, luteolin 7-b-D-gluco- SF162) with IC50 values of 140 and 40 nM, 144 nM and 1 mM,
pyranosyl-200 -sulfate, named thalassiolin A (148), was isolated and 189 nM and 1.3 mM, respectively. They also inhibited HIV-1-
from the Caribbean sea grass Thalassia testudinum. It inhibited mediated cell fusion against LAV, with IC50 values of 41 nM, 1.3
HIV-1 integrase terminal cleavage and strand transfer activities and 3.9 mM, respectively. These results suggested that these
with IC50 values of 2.1 and 0.4 mM, respectively, and also showed compounds might act at the early stages of HIV-1 entry. Mir-
antiviral activity in an MT-2 cell based assay, with an IC50 of abamide A (156), which contains the rhamnosylated b-OMe Tyr
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F

27 mM. Molecular docking studies in HIV-1 IN suggested that residue, was observed to be the most potent of all the isolated
148 can be accommodated near the catalytic site of the enzyme, compounds. Earlier, it had been suggested that the b-OMe Tyr
the binding mode overlapping with the location of the ligand residue is critical for the anti-HIV activity of a structurally
(5CITEP). The tetrazole ring of 5CITEP occupies the same similar compound, theopapuamide.107 However, it was found
binding position as the sulfated glucose of 148, and the orien- that in the case of mirabamides, the b-OMe Tyr was not the only
Downloaded by University of Sussex on 06 August 2012

tations of the keto–enol and chloroindole moieties of 5CITEP essential residue for activity, because b-OMe Tyr with substitu-
show similarities with the benzopyranone binding position.98 tion at the 40 -position can be tolerated with no adverse effects on
Diphlorethohydroxycarmalol (149), a carmalol derivative antiviral activity, and the presence of a free 40 -hydroxyl on the
isolated from the marine brown alga Ishige okamurae collected Tyr unit was not essential. Yet another important finding was
from the coasts of Korea, exhibited inhibitory effects on HIV-1 that compound 156 was equipotent to papuamide A (which
reverse transcriptase and integrase, with IC50 values of 9.1 mM contains a b-OMe Tyr), while theopapauamide (which lacks
and 25.2 mM, respectively. However, it did not show any inhib- a b-OMe Tyr residue) was inactive. The b-OMe Tyr residue
itory activity against HIV-1 PR.99 Phloroglucinol derivative 6,60 - provides a specific conformation required for binding to the
bieckol (150) was isolated from Korean brown alga Ecklonia target protein involved in HIV-1 entry.108 Homophymine A
cava, and is naturally abundant in the genus. Kim et al. showed (159), a cyclodepsipeptide isolated from the marine sponge
that 150 had inhibitory activity against HIV-1-induced syncytia Homophymia sp. collected in New Caledonia, effectively inhibi-
formation (EC50 1.72 mM), lytic effects (EC50 1.23 mM), and viral ted HIV-1IIIb infection with an IC50 of 75 nM. Direct cytotoxicity
p24 antigen production (EC50 1.26 mM). It also selectively of 159 against the host cells was observed, with a TC50 of
inhibited the activity of HIV-1 RT, with an EC50 of 1.07 mM.100 1.19 mM.109 The structures are shown in Fig. 12.
Two diterpenes, (6R)-6-hydroxydichotoma-3,14-diene-1,17-
dial (151) and (6R)-6-acetoxydichotoma-3,14-diene-1,17-dial
(152) were isolated from the Brazilian brown alga Dictyota
5 Advanced-stage molecules – where are they now?
menstrualis. Compounds 151 and 152 inhibited virus replication
with EC50 of 40 and 70 mM, respectively. Moreover, 151 and 152 In the past, several anti-HIV natural products have proceeded to
reduced HIV-1 RT activity, with IC50 values of 10 and 35 mM,101 various stages of drug development. The structures of these
and specifically inhibited RNA-dependent DNA polymerase, molecules is given in Fig. 13, and their present status is provided
with Ki values of 10 and 35 mM. Neither compound inhibited in Table 1.
DNA-dependent DNA-polymerase102 or affected cell viability or Calanolide was the most active anti-HIV-1 compound isolated
proliferation, and therefore the synthesis of proviral DNA was in the NCI-led discovery program from the rainforest tree
considered as a specific target for the anti-HIV-1 activity of these Calophyllum lanigerum.110 (+)-Calanolide A (160) inhibited
diterpenes.101 various laboratory strains of HIV-1 with EC50 0.10–0.17 mM. It
Dolabellane diterpene 8,10,18-trihydroxy-2,6-dolabelladiene selectively inhibited recombinant HIV-1 RT, but not cellular
(153) isolated from the alga Dictyota pfaffii, inhibited HIV-1 DNA polymerases or HIV-2 RT.111 Mechanism-of-action studies
infection in PBMCs, with an EC50 of 8.4 mM, and in macro- revealed that it bound near the active site of the enzyme HIV-1
phages, with an EC50 of 1.7 mM. It also showed inhibition of RT and interfered with dNTP binding. It inhibited HIV-1 RT in
HIV-1 RT, with an IC50 of 16.5 mM.103,104 a synergistic manner with nevirapine, suggesting that it was
A new furanoterpene, designated dehydrofurodendin, was different than the general class of nucleoside RT inhibitors.112 A
isolated from Madagascan sponges of the genus Lendenfeldia. single amino acid change in HIV-1 RT (V106A or Y181C) lead to
Dehydrofurodendin (154) inhibited HIV-1 RT-associated DNA significant resistance to nevirapine, but sensitivity to calanolide
polymerase activities, with IC50 values of 3.2 and 5.6 mM for A was unaffected. In contrast, the Y188H mutant had 30-fold
RNA- and DNA-directed DNA polymerase functions, respec- increased resistance to calanolide A, while nevirapine did not
tively. It also inhibited the RNase H activity of HIV-1 RT, with show any resistance.113
an IC50 value of 29.5 mM.105 The efficacy of 160 was demonstrated in vivo in a hollow-fiber
Neamphamide A (155), a cyclic depsipeptide containing 11 mouse model following oral or parenteral administration.114 The
amino acid residues, was isolated from a Papua New Guinea pharmacokinetic data after i.v. administration of calanolide A
collection of the marine sponge Neamphius huxleyi, and was and dihydrocalanolide A were almost the same (AUC (area

1794 | Nat. Prod. Rep., 2010, 27, 1781–1800 This journal is ª The Royal Society of Chemistry 2010
View Online
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F
Downloaded by University of Sussex on 06 August 2012

Fig. 12 Anti-HIV compounds of marine origin.

under the curve), t1/2, clearance). Dihydrocalanolide A showed taste perversion, headache, eructation, and nausea.116,117 The
better oral bioavailability.115 latest information available states that (+)-calanolide A is in
Phase I studies in 47 healthy volunteers using 160 encapsulated Phase IB trials for evaluation of the safety, pharmacokinetics,
in a sesame oil-based soft gelatin capsule demonstrated its safety and effects in HIV-positive patients.118
and favorable pharmacokinetic profile. No serious adverse side- Bevirimat, 3-O-(30 ,30 -dimethylsuccinyl)betulinic acid (161),
effects were reported; the majority of all adverse events were showed a potent activity in acutely infected H9 lymphocytes with
ranked as grade 1 (mild), and included symptoms of dizziness, an EC50 value of <0.35 nM.119 The design of 161 was based on

This journal is ª The Royal Society of Chemistry 2010 Nat. Prod. Rep., 2010, 27, 1781–1800 | 1795
View Online
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F

Fig. 13 Advanced-stage molecules.


Downloaded by University of Sussex on 06 August 2012

Table 1 Advanced-stage anti-HIV natural products DCK, 30 R,40 R-di-O-()-camphanoyl-(+)-cis-khellactone


(162), has been synthesized from suksdorfin by replacing the 30 -
Molecule Stage acetyl and 40 -isovaleryl groups with camphanoyl esters. It
Calanolide A Phase I showed potent activity against HIV-1IIIB compared to suksdor-
Bevirimat Phase IIb fin, with an EC50 of 0.049 mM.126 SAR studies suggested that
PA-1050040 Phase I a methyl group at C-3, C-4, or C-5 increased anti-HIV activity.
DCK Preclinical 4-Methyl-DCK was 10 times more active than DCK (EC50
DCQA Preclinical
Dextran sulfate Phase I 0.0059 mM).127 Preliminary studies on the mechanism of action
Curcumin Preclinical showed that it selectively inhibited virus replication in HIV-1-
Cyanovirin A Preclinical infected cells, but did not inhibit the activity of three major HIV
enzymes, HIV-RT, PR, and IN. The studies indicated that 162
inhibited HIV after viral entry but prior to viral DNA integra-
HIV-1 entry inhibitors with the same skeleton (betulinic acid). tion.4 DCK inhibits HIV-RT at a later step than that affected by
Bevirimat did not affect viral attachment or fusion, RT, PR or AZT and other clinically approved RT inhibitors. Studies on the
IN function, but instead acted at a late stage in HIV replica- analog hydroxymethyl DCK suggested that it inhibited the
tion.120 Studies to understand its mechanism of action revealed production of double-stranded viral DNA from the single-
that 161 dose-dependently inhibited the processing of the viral stranded DNA intermediate.120 The novel mechanism of action
Gag polyprotein at a specific step, namely the conversion of the of DCK is known as strand-transfer inhibition.128 Several new
capsid precursor p25 to mature capsid protein p24.121 This analogs of DCK have been found to be more active.4,120
inhibition led to the production of non-infectious and immature Several dicaffeoylquinic acids (DCQAs) were found to inhibit
viral particles, resulting in a new class of anti-HIV agents known HIV-1 replication at 1–6 mM in T-cell lines, whereas they were
as maturation inhibitors.66 Based on the potent in vitro and in non-cytotoxic up to 120 mM. A mechanism-based study sug-
vivo antiviral activity along with supportive preclinical data, 161 gested that the compounds inhibited HIV-1 IN. Molecular
was advanced into clinical testing and was granted ‘Fast-Track’ modelling of DCQAs with the core catalytic domain of HIV-1
New Drug Development status by the US FDA in December IN indicated that the inhibitors occupy a groove within the
2004.120 As of 2008, it was in Phase IIb clinical trials.66 Bevirimat catalytic site.129 1,5-DCQA (163), another member of the
offers a favorable pharmacokinetic profile in healthy humans DCQA group, was found to have low bioavailability and
and HIV-infected patients after a single oral administration.122,123 remained intact in rat plasma following oral administration.
Bevirimat was well absorbed after oral administration, and peak Twenty-two metabolites were found in the matrices of rats,
plasma concentrations were achieved in 1–3 h. A long half-life of suggesting that the extensive metabolism might be one factor
60–80 h may facilitate once-daily dosing. It is not oxidatively leading to the low bioavailability of 163. Three major metabo-
metabolized, does not inhibit cytochrome P450,122 and is elimi- lites of 163 also inhibited HIV replication in MT-4 cells, with
nated by hepatic glucuronidation.124 IC50 values of 25, 25 and 46 mM respectively, making an
PA1050040, an analog of bevirimat, acts as a maturation important contribution to the anti-HIV activity.130 In another
inhibitor and is a potent inhibitor of virus replication in primary study, following a single intravenous administration of 163, it
and prototypic HIV-1 isolates, with an EC50 of 15 nM. It was disappeared rapidly from plasma with a mean terminal half-life
also active against the bevirimat-resistant wild-type isolate (t1/2) of 1.40 h. A total of 15 metabolites along with the parent
L363M, and isolates resistant to approved ARVs. It is orally molecule were observed in rat urine. The quick urinary excre-
bioavailable in rats. The latest information available indicates tion and extensive metabolism of 163 result in its rapid elimi-
that it has entered Phase I clinical trials for single-dose testing.125 nation from the system.131

1796 | Nat. Prod. Rep., 2010, 27, 1781–1800 This journal is ª The Royal Society of Chemistry 2010
View Online

The sulfated polysaccharide dextran sulfate inhibited the repli- targeting specific steps in the viral life-cycle. In this review, we
cation of HIV-1 with an IC50 of 9.1 mg/mL in MT-4 cells. Dextran have provided an overview of various natural products isolated
sulfates with molecular weights of 5000, 8000 and 500 000 showed from diverse sources and acting at different targets/steps of the
a similar inhibitory effect on HIV-1 replication. They also showed life-cycle, dissecting the work done over the last seven years into
an HIV-1-RT inhibitory effect, with IC50 values of 41.9, 66.2 and two broad categories, i.e. new natural products showing anti-
17.9 mg/mL, respectively, and interfere with virus adsorption.132 HIV activity and previously known natural products that were
These sulfated polysaccharides are also potent inhibitors of other evaluated for this activity. As can be observed, during the period
enveloped viruses such as HSV, cytomegalovirus and vesicular covered in this review, fewer new natural products were discov-
stomatitis virus,133 and also show strong anti-coagulant proper- ered as having anti-HIV potential compared to the much larger
ties.132 Unfortunately, clinical trials of oral dextran sulfate in HIV- number of previously known natural products found to be active
infected patients were terminated because of poor absorption of the in various assays to assess anti-HIV potential. Therefore,
drug, poor central nervous system penetration and short half-life.134 a multidirectional approach needs to be continued for discovery
Furthermore, parenteral dextran sulfate did not lower circulating of anti-HIV drugs from natural sources. This approach should
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F

HIV antigen levels, but instead increased the circulating antigen. include finding new natural products, evaluating known natural
The dextran sulfate infusion was also associated with significant products having structural similarity with known anti-HIV
toxicity, especially thrombocytopenia. As a result, further clinical compounds, semi-synthetic modifications of anti-HIV natural
development of dextran sulfate as a therapy for symptomatic HIV products in order to enhance their activity, and preparation of
infection was not warranted, and the authors advised caution in the composite extracts active in anti-HIV assays. Here, it becomes
Downloaded by University of Sussex on 06 August 2012

clinical evaluation of other polysulfated polyanions.135 important to emphasize the development of alternative therapies
Curcumin (164) is the yellow pigment of turmeric (Curcuma using medicinal plants showing anti-HIV activities. Extracts
longa), a widely used spice and food coloring agent. Curcumin is from plants showing anti-HIV activity should be profiled using
reported to show antitumor, anti-inflammatory and many other techniques such as HPLC–PDA and LC–MS for complete che-
biological activities. It also plays a role in the prevention of moprofiling to give standardized extracts that can be used as
neurological, cardiovascular and metabolic disorders.136 Curcu- herbal products. Natural products have served as important
min showed HIV-1 IN inhibition, with an IC50 of 40 mM for strand leads for the discovery of more potent molecules – for example,
transfer reaction. HIV-1 IN inhibition by curcumin may result bevirimat and DCK were designed on the basis of anti-HIV
from coordination of its hydroxyl groups to the acidic residues in activity of naturally occurring betulinic acid and suksdorfin,
the active site, but it was suggested that the inhibition may involve respectively. The possibility of finding new molecules based on
multiple mechanisms.137 It was also reported that it inhibits p24 natural products acting in the nanomolar/picomolar range need
antigen production and Tat-mediated transcription at higher to be explored to find effective drugs against HIV. Although very
concentration.138 There have been almost 2500 in vitro and in vivo few natural products (such as clusianone, propolone A, the lectin
studies on curcumin for diverse activities, and this has generated BanLec, and mirabamide A) have shown anti-HIV activity in the
enough interest for clinical trials in cancer and inflammation. One nanomolar/picomolar range, further efforts are warranted to find
of the most important limitations of curcumin is its low bioavail- new lead molecules for HIV/AIDS from natural sources.
ability, which is due to its hydrophobic nature, and numerous
approaches have been undertaken to enhance its bioavailability.136
The safety profile and pharmacokinetic data are well established, 7 Acknowledgement
and it is currently in clinical trials for AIDS patients.137 We thank the Director of NIPER for support.
Cyanovirin-N, an 11 kDa protein containing 101 amino acids,
was isolated from extracts of the cultured cyanobacterium
Nostoc ellipsosporum. It irreversibly inactivates laboratory 8 References
strains and primary isolates of HIV-1 and HIV-2 in low nano-
1 AIDS Epidemic Update 2009, UNAIDS, Geneva, Switzerland, 2009.
molar concentrations. It also inhibits cell-to-cell fusion and 2 2008 Report on the global AIDS epidemic, UNAIDS, Geneva,
transmission of HIV-1 infection.139 Cyanovirin-N binds to viral Switzerland, 2008.
envelope glycoproteins, and interferes with viral interactions 3 W. C. Greene, Z. Debyser, Y. Ikeda, E. O. Freed, E. Stephens,
with target cell receptors that are essential for viral entry and cell- W. Yonemoto, R. W. Buckheit, J. A. Esteg and T. Cihlar,
Antiviral Res., 2008, 80, 251–265.
to-cell fusion, targeting N-linked high-mannose oligosaccharides 4 D. Yu, M. Suzuki, L. Xie, S. L. Morris-Natschke and K. H. Lee,
on the viral envelope.140 Intravenous administration of cyano- Med. Res. Rev., 2003, 23, 322–345.
virin-N resulted in rapid removal by renal filtration. It has a high 5 R. S. B. Goncalves, M. L. Ferreira, M. T. Ventura, M. A. Peralta
and S. M. V. N. De, Rev. Fitos, 2006, 2, 74–81.
potential for use as a systemic compound to control viral load or
6 J. Nian and J. Dong, Zhongguo Yaoxue Zazhi, 2005, 40, 571–573.
in the development of microbiocides to prevent primary viral 7 S. B. Singh, F. Pelaez, D. J. Hazuda and R. B. Lingham, Drugs
infection. It was suggested that PEGylated cyanovirin-N would Future, 2005, 30, 277–299.
be helpful to overcome problems associated with the use of 8 G. Dong, Y. Li, Y. Liu and G. Su, Zhongguo Haiyang Yaowu, 2009,
28, 44–53.
proteins. These formulations are presently in preclinical phase.141 9 P. Cos, L. Maes, A. Vlietinck and L. Pieters, Planta Med., 2008, 74,
1323–1337.
10 J. Wang, Y. Zheng, T. Efferth, R. Wang, Y. Shen and X. Hao,
6 Conclusions and future perspectives Phytochemistry, 2005, 66, 697–701.
11 M. Horiuch, C. Murakami, N. Fukamiya, D. Yu, T. H. Chen,
The basic biological processes in the HIV life-cycle are now well- K. F. Bastow, D. C. Zhang, Y. Takaishi, Y. Imakura and
established, and it is possible to find natural compounds K. H. Lee, J. Nat. Prod., 2006, 69, 1271–1274.

This journal is ª The Royal Society of Chemistry 2010 Nat. Prod. Rep., 2010, 27, 1781–1800 | 1797
View Online

12 H. Jayasuriya, K. B. Herath, J. G. Ondeyka, J. D. Polishook, 42 W. L. Xiao, S. X. Huang, R. R. Wang, J. L. Zhong, X. M. Gao,


G. F. Bills, A. W. Dombrowski, M. S. Springer, S. Siciliano, F. He, J. X. Pu, Y. Lu, Y. T. Zheng, Q. T. Zheng and H. D. Sun,
L. Malkowitz, M. Sanchez, Z. Guan, S. Tiwari, D. W. Stevenson, Phytochemistry, 2008, 69, 2862–2866.
R. P. Borris and S. B. Singh, J. Nat. Prod., 2004, 67, 1036–1038. 43 X. M. Zhang, D. F. Chen, X. J. He, S. Yang, P. Zheng and
13 G. Li, K. M. Haney, G. E. Kellogg and Y. Zhang, J. Chem. Inf. M. H. Jiang, Acta Pharmacol. Sinica, 2000, 21, 373–376.
Model., 2009, 49, 120–132. 44 J. Zhang, L. L. Shi and Y. N. Zheng, Nat. Prod. Commun., 2010, 5,
14 P. W. Hsieh, F. R. Chang, K. H. Lee, T. L. Hwang, S. M. Chang and 231–234.
Y. C. Wu, J. Nat. Prod., 2004, 67, 1175–1177. 45 S. Y. Oh, Y. H. Kim, D. S. Bae, B. H. Um, C. H. Pan, C. Y. Kim,
15 E. M. Silva, C. C. Cirne-Santos, I. C. Frugulhetti, B. Galvao-Castro, H. J. Lee and J. K. Lee, Biosci., Biotechnol., Biochem., 2010, 74,
E. M. Saraiva, M. E. Kuehne and D. C. Bou-Habib, Planta Med., 285–291.
2004, 70, 808–812. 46 X. M. Gao, J. X. Pu, S. X. Huang, L. M. Yang, H. Huang,
16 W. Zhang, Y. Ramamoorthy, R. F. Tyndale, S. D. Glick, W. L. Xiao, Y. T. Zheng and H. D. Sun, J. Nat. Prod., 2008, 71,
I. M. Maisonneuve, M. E. Kuehne and E. M. Sellers, Drug Metab. 558–563.
Dispos., 2002, 30, 663–669. 47 Y. B. Yu, H. Miyashiro, N. Nakamura, M. Hattori and J. C. Park,
17 A. K. Chakravarty, T. Sarkar, K. Masuda and K. Shiojima, Arch. Pharmacal Res., 2007, 30, 820–826.
Phytochemistry, 1999, 50, 1263–1266. 48 J. Lee, W. K. Oh, J. S. Ahn, Y. H. Kim, J. T. Mbafor, J. Wandji and
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F

18 C. Ito, M. Itoigawa, A. Sato, C. M. Hasan, M. A. Rashid, Z. T. Fomum, Planta Med., 2009, 75, 268–270.
H. Tokuda, T. Mukainaka, H. Nishino and H. Furukawa, J. Nat. 49 C. H. Ma, R. R. Wang, R. R. Tian, G. Ye, M. S. Fan, Y. T. Zheng
Prod., 2004, 67, 1488–1491. and C. G. Huang, Chem. Nat. Compd., 2009, 45, 282–285.
19 B. Kongkathip, N. Kongkathip, A. Sunthitikawinsakul, 50 S. Tewtrakul, S. Subhadhirasakul, S. Cheenpracha and C. Karalai,
C. Napaswat and C. Yoosook, Phytother. Res., 2005, 19, 728–731. Phytother. Res., 2007, 21, 1092–1095.
20 M. J. Cheng, K. H. Lee, I. L. Tsai and I. S. Chen, Bioorg. Med. 51 C. C. Li, Z. X. Xie, Y. D. Zhang, J. H. Chen and Z. Yang, J. Org.
Chem., 2005, 13, 5915–5920. Chem., 2003, 68, 8500–8504.
Downloaded by University of Sussex on 06 August 2012

21 R. Stadler, T. M. Kutchan and M. H. Zenk, Phytochemistry, 1989, 52 S. Cheenpracha, C. Karalai, C. Ponglimanont, S. Subhadhirasakul
28, 1083–1086. and S. Tewtrakul, Bioorg. Med. Chem., 2006, 14, 1710–1714.
22 N. Samanani, D. K. Liscombe and P. J. Facchini, Plant J., 2004, 40, 53 V. Martino, J. Morales, J. J. Martinez-Irujo, M. Font, A. Monge
302–313. and J. Coussio, Phytother. Res., 2004, 18, 667–669.
23 Y. Kashiwada, A. Aoshima, Y. Ikeshiro, Y. P. Chen, H. Furukawa, 54 C. C. Lin, Y. F. Hsu, T. C. Lin and H. Y. Hsu, Phytother. Res., 2001,
M. Itoigawa, T. Fujioka, K. Mihashi, L. M. Cosentino, S. L. Morris- 15, 206–212.
Natschke and K. H. Lee, Bioorg. Med. Chem., 2005, 13, 443–448. 55 S. Jain, P. P. Yadav, V. Gill, N. Vasudeva and N. Singla, Phytochem.
24 M. H. Yan, P. Cheng, Z. Y. Jiang, Y. B. Ma, X. M. Zhang, Rev., 2009, 8, 491–502.
F. X. Zhang, L. M. Yang, Y. T. Zheng and J. J. Chen, J. Nat. 56 M. K. Reddy, S. K. Gupta, M. R. Jacob, S. I. Khan and D. Ferreira,
Prod., 2008, 71, 760–763. Planta Med., 2007, 73, 461–467.
25 A. L. Otshudi, S. Apers, L. Pieters, M. Claeys, C. Pannecouque, 57 J. L. Lopez-Perez, D. A. Olmedo, E. del Olmo, Y. Vasquez,
E. De Clercq, A. Van Zeebroeck, S. Lauwers, M. Frederich and P. N. Solis, M. P. Gupta and A. San Feliciano, J. Nat. Prod.,
A. Foriers, J. Ethnopharmacol., 2005, 102, 89–94. 2005, 68, 369–373.
26 M. L. Lohombo-Ekomba, P. N. Okusa, O. Penge, C. Kabongo, 58 N. Marquez, R. Sancho, L. M. Bedoya, J. Alcami, J. L. Lopez-Perez,
M. I. Choudhary and O. E. Kasende, J. Ethnopharmacol., 2004, A. S. Feliciano, B. L. Fiebich and E. Munoz, Antiviral Res., 2005, 66,
93, 331–335. 137–145.
27 L. Szlavik, A. Gyuris, J. Minarovits, P. Forgo, J. Molnar and 59 L. M. Bedoya, M. Beltran, R. Sancho, D. A. Olmedo, S. Sanchez-
J. Hohmann, Planta Med., 2004, 70, 871–873. Palomino, E. del Olmo, J. L. L opez-Perez, E. Mu~ noz,
28 B. Sener, I. Orhan and J. Satayavivad, Phytother. Res., 2003, 17, A. S. Feliciano and J. Alcamı, Bioorg. Med. Chem. Lett., 2005, 15,
1220–1223. 4447–4450.
29 P. L. Wu, F. W. Lin, T. S. Wu, C. S. Kuoh, K. H. Lee and S. J. Lee, 60 V. Reutrakul, N. Anantachoke, M. Pohmakotr, T. Jaipetch,
Chem. Pharm. Bull., 2004, 52, 345–349. S. Sophasan, C. Yoosook, J. Kasisit, C. Napaswat, T. Santisuk
30 A. Saklani and S. K. Kutty, Drug Discovery Today, 2008, 13, 161– and P. Tuchinda, Planta Med., 2007, 73, 33–40.
171. 61 V. Reutrakul, W. Chanakul, M. Pohmakotr, T. Jaipetch,
31 M. Chen, N. Kilgore, K. H. Lee and D. F. Chen, J. Nat. Prod., 2006, C. Yoosook, J. Kasisit, C. Napaswat, T. Santisuk, S. Prabpai,
69, 1697–1701. P. Kongsaeree and P. Tuchinda, Planta Med., 2006, 72, 1433–1435.
32 Q. Z. Sun, D. F. Chen, P. L. Ding, C. M. Ma, H. Kakuda, 62 A. L. Piccinelli, O. Cuesta-Rubio, M. B. Chica, N. Mahmood,
N. Nakamura and M. Hattori, Chem. Pharm. Bull., 2006, 54, 129–132. B. Pagano, M. Pavone, V. Barone and L. Rastrelli, Tetrahedron,
33 X. N. Li, J. X. Pu, X. Du, L. M. Yang, H. M. An, C. Lei, F. He, 2005, 61, 8206–8211.
X. Luo, Y. T. Zheng, Y. Lu, W. L. Xiao and H. D. Sun, J. Nat. 63 N. M. Ahmad, V. Rodeschini, N. S. Simpkins, S. E. Ward and
Prod., 2009, 72, 1133–1141. A. J. Blake, J. Org. Chem., 2007, 72, 4803–4815.
34 K. M. Meragelman, T. C. McKee and M. R. Boyd, J. Nat. Prod., 64 L. Verotta, Phytochem. Rev., 2002, 1, 389–407.
2001, 64, 546–548. 65 J. S. Lee, M. Hattori and J. Kim, Planta Med., 2008, 74, 532–534.
35 T. C. McKee, H. R. Bokesch, J. L. McCormick, M. A. Rashid, 66 R. Y. Kuo, K. Qian, S. L. Morris-Natschke and K. H. Lee, Nat.
D. Spielvogel, K. R. Gustafson, M. M. Alavanja, J. H. Cardellina Prod. Rep., 2009, 26, 1321–1344.
and M. R. Boyd, J. Nat. Prod., 1997, 60, 431–438. 67 Y. Wei, C.-M. Ma and M. Hattori, Phytochem. Lett., 2009, 2, 63–66.
36 V. Reutrakul, N. Ningnuek, M. Pohmakotr, C. Yoosook, 68 V. Reutrakul, N. Anantachoke, M. Pohmakotr, T. Jaipetch,
C. Napaswad, J. Kasisit, T. Santisuk and P. Tuchinda, Planta C. Yoosook, J. Kasisit, C. Napaswa, A. Panthong, T. Santisuk,
Med., 2007, 73, 683–688. S. Prabpai, P. Kongsaeree and P. Tuchinda, Planta Med., 2010,
37 S. Cao, C. Rossant, S. Ng, A. D. Buss and M. S. Butler, 76, 368–371.
Phytochemistry, 2003, 64, 987–990. 69 X. Luo, Y. Chang, X. J. Zhang, J. X. Pu, X. M. Gao, Y. L. Wu,
38 G. Appendino, M. Ottino, N. Marquez, F. Bianchi, A. Giana, R. R. Wang, W. L. Xiao, Y. T. Zheng and Y. Lu, Tetrahedron
M. Ballero, O. Sterner, B. L. Fiebich and E. Munoz, J. Nat. Prod., Lett., 2009, 50, 5962–5964.
2007, 70, 608–612. 70 M. Daoubi, N. Marquez, N. Mazoir, A. Benharref, R. Hernandez-
39 N. T. Dat, K. Bae, A. Wamiru, J. B. McMahon, S. F. Le Grice, Galan, E. Munoz and I. G. Collado, Bioorg. Med. Chem., 2007,
M. Bona, J. A. Beutler and Y. H. Kim, J. Nat. Prod., 2007, 70, 15, 4577–4584.
839–841. 71 H. J. Zhang, V. H. Nguyen, M. C. Nguyen, D. D. Soejarto,
40 H. J. Kim, E. J. Kim, S. H. Seo, C. G. Shin, C. Jin and Y. S. Lee, J. J. M. Pezzuto, H. H. Fong and G. T. Tan, Planta Med., 2005, 71,
Nat. Prod., 2006, 69, 600–603. 452–457.
41 J. X. Pu, L. M. Yang, W. L. Xiao, R. T. Li, C. Lei, X. M. Gao, 72 R. R. Tian, J. C. Chen, G. H. Zhang, M. H. Qiu, Y. H. Wang, L. Du,
S. X. Huang, S. H. Li, Y. T. Zheng, H. Huang and H. D. Sun, X. Shen, N. F. Liu and Y. T. Zheng, Chin. J. Nat. Med., 2008, 6,
Phytochemistry, 2008, 69, 1266–1272. 214–218.

1798 | Nat. Prod. Rep., 2010, 27, 1781–1800 This journal is ª The Royal Society of Chemistry 2010
View Online

73 W. Song, Y. Ma, X. Bai, X. Zhang, Q. Gu, Y. Zheng, J. Zhou and 104 C. C. Cirne-Santos, V. L. Teixeira, L. R. Castello-Branco,
J. Chen, Planta Med., 2007, 73, 372–375. I. C. Frugulhetti and D. C. Bou-Habib, Planta Med., 2006, 72,
74 H. S. Bodiwala, S. Sabde, D. Mitra, K. K. Bhutani and I. P. Singh, 295–299.
Nat. Prod. Commun., 2009, 4, 1173–1175. 105 L. Chill, A. Rudi, M. Aknin, S. Loya, A. Hizi and Y. Kashman,
75 W. Woradulayapinij, N. Soonthornchareonnon and C. Wiwat, Tetrahedron, 2004, 60, 10619–10626.
J. Ethnopharmacol., 2005, 101, 84–89. 106 N. Oku, K. R. Gustafson, L. K. Cartner, J. A. Wilson,
76 X. Ye and T. B. Ng, J. Agric. Food Chem., 2009, 57, 4366–4371. N. Shigematsu, S. Hess, L. K. Pannell, M. R. Boyd and
77 S. K. Lam and T. B. Ng, Phytomedicine, 2009, 16, 444–450. J. B. McMahon, J. Nat. Prod., 2004, 67, 1407–1411.
78 T. Mori, B. R. O’Keefe, R. C. Sowder, 2nd, S. Bringans, R. Gardella, 107 A. S. Ratnayake, T. S. Bugni, X. Feng, M. K. Harper, J. J. Skalicky,
S. Berg, P. Cochran, J. A. Turpin, R. W. Buckheit, Jr., J. B. McMahon K. A. Mohammed, C. D. Andjelic, L. R. Barrows and C. M. Ireland,
and M. R. Boyd, J. Biol. Chem., 2005, 280, 9345–9353. J. Nat. Prod., 2006, 69, 1582–1586.
79 C. K. Wang, M. L. Colgrave, K. R. Gustafson, D. C. Ireland, 108 A. Plaza, E. Gustchina, H. L. Baker, M. Kelly and C. A. Bewley,
U. Goransson and D. J. Craik, J. Nat. Prod., 2008, 71, 47–52. J. Nat. Prod., 2007, 70, 1753–1760.
80 S. Rechter, T. K€ onig, S. Auerochs, S. Thulke, H. Walter, 109 A. Zampella, V. Sepe, P. Luciano, F. Bellotta, M. C. Monti,
H. D€ ornenburg, C. Walter and M. Marschall, Antiviral Res., 2006, M. V. D’Auria, T. Jepsen, S. Petek, M. T. Adeline, O. Laprevote,
72, 197–206. A. M. Aubertin, C. Debitus, C. Poupat and A. Ahond, J. Org.
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F

81 P. Lin and T. B. Ng, J. Agric. Food Chem., 2008, 56, 10481–10486. Chem., 2008, 73, 5319–5327.
82 J. Balzarini, S. Hatse, K. Vermeire, K. Princen, S. Aquaro, 110 Y. Kashman, K. R. Gustafson, R. W. Fuller, J. H. Cardellina,
C. F. Perno, E. De Clercq, H. Egberink, G. Vanden Mooter, J. B. McMahon, M. J. Currens, R. W. Buckheit, S. H. Hughes,
W. Peumans, E. Van Damme and D. Schols, Antimicrob. Agents G. M. Cragg and M. R. Boyd, J. Med. Chem., 1992, 35, 2735–2743.
Chemother., 2004, 48, 3858–3870. 111 M. J. Currens, R. J. Gulakowski, J. M. Mariner, R. A. Moran,
83 E. F. Fang, P. Lin, J. H. Wong, S. W. Tsao and T. B. Ng, J. Agric. R. W. Buckheit, K. R. Gustafson, J. B. McMahon and
Food Chem., 2010, 58, 2221–2229. M. R. Boyd, J. Pharmacol. Exp. Ther., 1996, 279, 645–651.
Downloaded by University of Sussex on 06 August 2012

84 M. D. Swanson, H. C. Winter, I. J. Goldstein and D. M. Markovitz, 112 M. J. Currens, J. M. Mariner, J. B. McMahon and M. R. Boyd,
J. Biol. Chem., 2010, 285, 8646–8655. J. Pharmacol. Exp. Ther., 1996, 279, 652–661.
85 Q. Gu, R. R. Wang, X. M. Zhang, Y. H. Wang, Y. T. Zheng, J. Zhou 113 Y. Quan, D. Motakis, R. Buckheit, Z. Q. Xu, M. T. Flavin,
and J. J. Chen, Planta Med., 2007, 73, 279–282. M. A. Parniak and M. A. Wainberg, Antiviral Ther., 1999, 4, 203–
86 R. R. Wang, Q. Gu, Y. H. Wang, X. M. Zhang, L. M. Yang, 209.
J. Zhou, J. J. Chen and Y. T. Zheng, J. Ethnopharmacol., 2008, 114 Z. Q. Xu, M. G. Hollingshead, S. Borgel, C. Elder, A. Khilevich and
117, 249–256. M. T. Flavin, Bioorg. Med. Chem. Lett., 1999, 9, 133–138.
87 Y. Zhang, R. Tian, S. Liu, X. Chen, X. Liu and Y. Che, Bioorg. Med. 115 R. A. Newman, W. Chen and T. L. Madden, J. Pharm. Sci., 1998,
Chem., 2008, 16, 2627–2634. 87, 1077–1080.
88 A. M. Haidle and A. G. Myers, Proc. Natl. Acad. Sci. U. S. A., 2004, 116 T. Creagh, J. L. Ruckle, D. T. Tolbert, J. Giltner, D. A. Eiznhamer,
101, 12048–12053. B. Dutta, M. T. Flavin and Z. Q. Xu, Antimicrob. Agents
89 L. Liu, S. Liu, X. Chen, L. Guo and Y. Che, Bioorg. Med. Chem., Chemother., 2001, 45, 1379–1386.
2009, 17, 606–613. 117 D. A. Eiznhamer, T. Creagh, J. L. Ruckle, D. T. Tolbert, J. Giltner,
90 K. Yoganathan, C. Rossant, S. Ng, Y. Huang, M. S. Butler and B. Dutta, M. T. Flavin, T. Jenta and Z. Q. Xu, HIV Clin. Trials,
A. D. Buss, J. Nat. Prod., 2003, 66, 1116–1117. 2002, 3, 435–450.
91 B. Z. Yu, G. H. Zhang, Z. Z. Du, Y. T. Zheng, J. C. Xu and 118 Anonymous, The safety and effectiveness of (+)-calanolide A in HIV-
X. D. Luo, Phytochemistry, 2008, 69, 2523–2526. infected patients who have never taken anti-HIV drugs, http://
92 X.-W. Zou, B.-D. Sun, X.-L. Chen, X.-Z. Liu and Y.-S. Che, Chin. J. clinicaltrial.gov/ct2/show/NCT00005120?term¼calanolide&rank ¼
Nat. Med., 2009, 7, 140–144. 1, Accessed 1 August, 2010.
93 S. W. Yang, R. Mierzwa, J. Terracciano, M. Patel, V. Gullo, 119 Y. Kashiwada, F. Hashimoto, L. M. Cosentino, C. H. Chen,
N. Wagner, B. Baroudy, M. Puar, T. M. Chan, A. T. McPhail and P. E. Garrett and K. H. Lee, J. Med. Chem., 1996, 39, 1016–1017.
M. Chu, J. Nat. Prod., 2006, 69, 1025–1028. 120 D. Yu, S. L. Morris-Natschke and K. H. Lee, Med. Res. Rev., 2007,
94 K. Yoganathan, C. Rossant, R. P. Glover, S. Cao, J. J. Vittal, S. Ng, 27, 108–132.
Y. Huang, A. D. Buss and M. S. Butler, J. Nat. Prod., 2004, 67, 121 F. Li, R. Goila-Gaur, K. Salzwedel, N. R. Kilgore, M. Reddick,
1681–1684. C. Matallana, A. Castillo, D. Zoumplis, D. E. Martin,
95 S. B. Singh, J. G. Ondeyka, N. Tsipouras, C. Ruby, V. Sardana, J. M. Orenstein, G. P. Allaway, E. O. Freed and C. T. Wild, Proc.
M. Schulman, M. Sanchez, F. Pelaez, M. W. Stahlhut, S. Munshi, Natl. Acad. Sci. U. S. A., 2003, 100, 13555–13560.
D. B. Olsen and R. B. Lingham, Biochem. Biophys. Res. Commun., 122 D. E. Martin, R. Blum, J. Wilton, J. Doto, H. Galbraith,
2004, 324, 108–113. G. L. Burgess, P. C. Smith and C. Ballow, Antimicrob. Agents
96 H. Jayasuriya, D. L. Zink, J. D. Polishook, G. F. Bills, Chemother., 2007, 51, 3063–3066.
A. W. Dombrowski, O. Genilloud, F. F. Pelaez, L. Herranz, 123 P. F. Smith, A. Ogundele, A. Forrest, J. Wilton, K. Salzwedel,
D. Quamina, R. B. Lingham, R. Danzeizen, P. L. Graham, J. Doto, G. P. Allaway and D. E. Martin, Antimicrob. Agents
J. E. Tomassini and S. B. Singh, Chem. Biodiversity, 2005, 2, 112– Chemother., 2007, 51, 3574–3581.
122. 124 D. E. Martin, K. Salzwedel and G. P. Allaway, Antiviral Chem.
97 Y. B. Kim, Y. H. Kim, J. Y. Park and S. K. Kim, Bioorg. Med. Chemother., 2008, 19, 107–113.
Chem. Lett., 2004, 14, 541–544. 125 N. Kilgore, M. Reddick, M. Zuiderhof, D. Stanley, T. Nitz,
98 D. C. Rowley, M. S. T. Hansen, D. Rhodes, C. A. Sotriffer, H. Ni, P. Bullock, G. Allaway and D. Martin, Characterization of
J. A. McCammon, F. D. Bushman and W. Fenical, Bioorg. Med. PA1050040, a second-generation HIV-1 maturation inhibitor, http://
Chem., 2002, 10, 3619–3625. www.panacos.com/product_4.htm, Accessed 1 August, 2010.
99 M. J. Ahn, K. D. Yoon, C. Y. Kim, J. H. Kim, C. G. Shin and 126 L. Xie, Y. Takeuchi, L. M. Cosentino and K. H. Lee, J. Med. Chem.,
J. Kim, Phytother. Res., 2006, 20, 711–713. 1999, 42, 2662–2672.
100 M. Artan, Y. Li, F. Karadeniz, S. H. Lee, M. M. Kim and S. K. Kim, 127 L. Xie, Y. Takeuchi, L. M. Cosentino and K. H. Lee, Bioorg. Med.
Bioorg. Med. Chem., 2008, 16, 7921–7926. Chem. Lett., 1998, 8, 2151–2156.
101 H. S. Pereira, L. R. Leao-Ferreira, N. Moussatche, V. L. Teixeira, 128 H. Itokawa, S. L. Morris-Natschke, T. Akiyama and K. H. Lee,
D. N. Cavalcanti, L. J. Costa, R. Diaz and I. C. Frugulhetti, J. Nat. Med., 2008, 62, 263–280.
Antiviral Res., 2004, 64, 69–76. 129 W. E. Robinson, Jr., M. Cordeiro, S. Abdel-Malek, Q. Jia,
102 H. de Souza Pereira, L. R. Leao-Ferreira, N. Moussatche, S. A. Chow, M. G. Reinecke and W. M. Mitchell, Mol.
V. L. Teixeira, D. N. Cavalcanti, L. J. da Costa, R. Diaz and Pharmacol., 1996, 50, 846–855.
I. C. Frugulhetti, Planta Med., 2005, 71, 1019–1024. 130 B. Yang, Z. Meng, J. Dong, L. Yan, L. Zou, Z. Tang and G. Dou,
103 J. P. Barbosa, R. C. Pereira, J. L. Abrantes, C. C. Cirne dos Santos, Drug Metab. Dispos., 2005, 33, 930–936.
M. A. Rebello, I. C. Frugulhetti and V. L. Texeira, Planta Med., 131 B. Yang, Z. Y. Meng, L. P. Yan, J. X. Dong, L. B. Zou, Z. M. Tang
2004, 70, 856–860. and G. F. Dou, J. Pharm. Biomed. Anal., 2006, 40, 417–422.

This journal is ª The Royal Society of Chemistry 2010 Nat. Prod. Rep., 2010, 27, 1781–1800 | 1799
View Online

132 M. Baba, R. Pauwels, J. Balzarini, J. Arnout, J. Desmyter and E. De 138 C. J. Li, L. J. Zhang, B. J. Dezube, C. S. Crumpacker and
Clercq, Proc. Natl. Acad. Sci. U. S. A., 1988, 85, 6132–6136. A. B. Pardee, Proc. Natl. Acad. Sci. U. S. A., 1993, 90, 1839–1842.
133 M. Baba, R. Snoeck, R. Pauwels and E. de Clercq, Antimicrob. 139 M. R. Boyd, K. R. Gustafson, J. B. McMahon, R. H. Shoemaker,
Agents Chemother., 1988, 32, 1742–1745. B. R. O’Keefe, T. Mori, R. J. Gulakowski, L. Wu, M. I. Rivera,
134 K. J. Lorentsen, C. W. Hendrix, J. M. Collins, D. M. Kornhauser, C. M. Laurencot, M. J. Currens, J. H. Cardellina, 2nd,
B. G. Petty, R. W. Klecker, C. Flexner, R. H. Eckel and R. W. Buckheit, Jr., P. L. Nara, L. K. Pannell, R. C. Sowder, 2nd
P. S. Lietman, Ann. Intern. Med., 1989, 111, 561–566. and L. E. Henderson, Antimicrob. Agents Chemother., 1997, 41,
135 C. Flexner et al., Antimicrob. Agents Chemother., 1991, 35, 2544–2550. 1521–1530.
136 B. B. Aggarwal and B. Sung, Trends Pharmacol. Sci., 2009, 30, 85– 140 A. J. Bolmstedt, B. R. O’Keefe, S. R. Shenoy, J. B. McMahon and
94. M. R. Boyd, Mol. Pharmacol., 2001, 59, 949–954.
137 A. Mazumder, K. Raghavan, J. Weinstein, K. W. Kohn and 141 H. Zappe, M. E. Snell and M. J. Bossard, Adv. Drug Delivery Rev.,
Y. Pommier, Biochem. Pharmacol., 1995, 49, 1165–1170. 2008, 60, 79–87.
Published on 25 October 2010 on http://pubs.rsc.org | doi:10.1039/C0NP00025F
Downloaded by University of Sussex on 06 August 2012

1800 | Nat. Prod. Rep., 2010, 27, 1781–1800 This journal is ª The Royal Society of Chemistry 2010

You might also like