Abstract
Medicinal plants are one of the most important sources of antiviral agents and lead compounds. Lignans are a large class of natural compounds comprising two phenyl propane units. Many of them have demonstrated biological activities, and some of them have even been developed as therapeutic drugs. In this review, 630 lignans, including those obtained from medicinal plants and their chemical derivatives, were systematically reviewed for their antiviral activity and mechanism of action. The compounds discussed herein were published in articles between 1998 and 2020. The articles were identified using both database searches (e.g., Web of Science, Pub Med and Scifinder) using key words such as: antiviral activity, antiviral effects, lignans, HBV, HCV, HIV, HPV, HSV, JEV, SARS-CoV, RSV and influenza A virus, and directed searches of scholarly publisher’s websites including ACS, Elsevier, Springer, Thieme, and Wiley. The compounds were classified on their structural characteristics as 1) arylnaphthalene lignans, 2) aryltetralin lignans, 3) dibenzylbutyrolactone lignans, 4) dibenzylbutane lignans, 5) tetrahydrofuranoid and tetrahydrofurofuranoid lignans, 6) benzofuran lignans, 7) neolignans, 8) dibenzocyclooctadiene lignans and homolignans, and 9) norlignans and other lignoids. Details on isolation and antiviral activities of the most active compounds within each class of lignan are discussed in detail, as are studies of synthetic lignans that provide structure–activity relationship information.
Keywords: Lignans, Medicinal plants, Antiviral, HBV, HSV, HIV
Introduction
A virus is a small infectious agent ranging from ~ 20 nm to 300 nm in diameter and contains mostly bundles of gene strands of either RNA or DNA as its genome. Viruses are not autonomous organisms and require the host cell environment and cellular factors for their propagation (Bekhit and Bekhit 2014). Due to the intracellular properties of viruses, it is often difficult to design a treatment that inhibits viral replication directly without adverse effects on the infected cells. (Zinser et al. 2018; Bar-On et al. 2018).
Viral infections can be categorized as chronic or acute based on the length of time the infections and symptoms last. Some of the viruses causing the most widespread and harmful chronic infections are hepatitis B virus (HBV), hepatitis C viruses (HCV), herpes simplex virus (HSV), human immunodeficiency virus (HIV), and human papilloma virus (HPV). HIV alone has caused approximately 33 million deaths since the virus was first discovered in 1981 (WHO 2021). Acute viral infections cause several of the most common, severe and rapid infections. This was painfully illustrated in the coronavirus disease 2019 (COVID-19) pandemic (Wu and McGoogan 2020). As of Mar 2021, there are over 123 million confirmed SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) infected cases worldwide and over 2.7 million COVID-19 deaths (Worldometer 2021). Annually, seasonal influenza epidemics usually cause 250,000–500,000 deaths. However, they could also cause far more severe damage to human lives. For example, the 1918 H1N1 Spanish flu killed an estimated 50 million people, making it one of the worst pandemics in recorded human history (Short et al. 2018). The viral outbreaks like the Ebola virus (EBOV) in West Africa in 2014 received extensive media coverage due to its epidemic potential and high mortality rates of up to 90% (Martinez et al. 2014).
Many compounds have been tested on various viruses for their antivirtal activity in the past few decades, and some novel antiviral compounds are currently in the process of either preclinical or clinical trials (Chaudhuri et al. 2018). Although public health measures and prophylactic vaccines should remain the most effective means for prevetion of virus infection, for many virues vaccines are either not available or are inefficient. In addition, drug resistance is becoming increasingly common due to the expanding numbers of antiviral drugs. Long-term treatment with antiviral nucleoside analogues can cause delayed and, at times, severe mitochondrial toxicity. Kidney injury associated with antiviral drug use involves diverse mechanisms affecting renal transporters and tubule cells (Hussain et al. 2017; Izzedine et al. 2005). Therefore, the ongoing need to fight viral infections requires continued partnerships between medicinal chemists and biomedical researchers to identify and develop novel antiviral agents that are highly efficacious and cost-effective for the management and control of viral infections when vaccines and standard therapies are unavailable or ineffective.
Globally, medicinal plants have been used in traditional health care systems since ancient times and are still the most important source of medicines for the vast majority of the world’s population (Pushpa et al. 2013). Many traditionally used medicinal plants display strong antiviral activities, and some of them are used to treat animals and people who suffer from viral infections. In 2006, polyphenon E®, a partially-purified extract of green tea (Camellia sinensis) was approved as the first ever botanical drug by the US FDA to treat genital warts caused by HPV. Furthermore, recent studies showing the antiviral potential of plant extracts against viral strains resistant to conventional antiviral agents have contested modern drug discovery practices (Akram et al. 2018). Therefore, exploring the natural antiviral constituents of medicinal plants has garnered widespread and increased interest.
During the last 30 years, numerous broad-based screening programs have been carried out throughout the world to evaluate the inhibitory effects of medicinal plants on several viruses using in vitro and in vivo assays. For example, the Boots drug company (Nottingham, England) screened 288 plants for anti-influenza activity, and 12 of these plants were discovered to be effective against influenza viruses (Mukhtar et al. 2008). In another study, a joint international drug discovery program evaluated the antiviral effects of 3,760 plants extracts in Vietnam and Laos, which led to the identification of more than 90 plant leads that showed anti-HIV and anti-bird flu virus activities (Zhang et al. 2016; Rumschlag-Booms et al. 2011).
Lignans, constituted by the union of two phenylpropane units, are a large class of plant secondary metabolites with diversified chemical structures with many being biologically active including the US FDA approved anticancer drugs etoposide and teniposide (Newman and Cragg 2020). They have been found rich in fruits, seeds and vegetables, and received widespread interest due to their various biological activities including antioxidant, antitumor, antibacterial, antiviral, insecticidal, fungistatic, estrogenic, and antiestrogenic activities (Teponno et al. 2016). Their activity against the coronavirus SARS 2003 was also reported to be prominent (Wen et al. 2007).
The first antiviral lignan reported is podophyllin, a toxic lignan obtained from Podophyllum peltatum with antiviral activity against papilloma virus in 1942 (Kaplan 1942). Charlton had reviewed 49 natural lignans with antiviral activity from 1942 to 1997 (Charlton 1998). In a recent review article, Cui et al. reported about 25 representative lignans that showed antiviral activities (Cui et al. 2020). However, more than 600 lignans have been reported for their antiviral effects since 1998, and many of them showed potent properties. It’s therefore necessary to present a systematic review to include these antiviral lignans.
The present review summarizes the natural antiviral lignans or derivatives from 1998 to 2020. The lignans will be classified in 9 categories according to their chemical structure classes. Their antiviral activities against HIV, HBV, HCV, SARS, herpes simplex virus (HSV), HPV, Ebola virus, influenza virus, vesicular stomatitis virus (VSV) and other viruses will be discussed in detail. The antiviral mechanism studies of lignans will also be included when they were reported.
Arylnaphthalene-type lignans
In arylnaphthalene lignans, a naphthalene ring-system is made with carbons 1–8 and 7′ and 8′. There have been numerous arylnaphthalene lignans discovered from natural sources. Among the most common structural motifs found in this class of lignans, as with all classes of lignans, are various substitutions on the aromatic rings, a lactone moiety formed from C-8, C-9, C-8′, and C-9′, and glycosides. Many lignans in this class showed significant and broad antiviral activities, warranting them for further chemical and biological approaches for antiviral drug development.
Between 1998 and 2020, 17 arylnaphthalene lignans were isolated from plants and tested for antiviral activity (1–13, 32, 152 and 153) (see Table 1 for source plants, antiviral activities, and references). Of particular interest due to their potent antiviral activity (< 1 μM), are 2 and 11. We found four studies on synthetic arylnaphthalene lignans which displayed significant antiviral activities and provided information about structure–activity relationships.
Table 1.
Source | Parts | Compounds | Activity | Activity value | References |
---|---|---|---|---|---|
Daphne acutiloba Rehd. (Thymelaeaceae, Yunnan, China) | Leaves and stems | 1 | HIV-1 | EC50 15.6 μM, TI 35.62 | Cao et al. (2010) |
Eucalyptus globoidea Blakely (Myrtaceae, Australia) | Buds | 2 | HIV integrase | EC50 0.64 μM | Ovenden et al. (2004) |
Iryanthera megistophylla A. C. Sm. (Myristicaceae, Colombia) | Bark | 3 | HSV-1 | EC50 > 1150 μM | Ming et al. (2002) |
Phyllanthus acutissima Miq. (Euphorbiaceae, Thailand) | Aerial parts | 4,5 | HIV-1 | 4 EC50 < 7.4 μM | Tuchinda et al. (2008) |
Phyllanthus flexuosus (Sieb. Et Zucc.) Muell. Arg (Euphorbiaceac, Yunnan, China) | Roots | 6–10 | HSV-1 | inactive, EC50 value unavailable | Zhao et al. (2014) |
Justicia procumbens var. leucantha (Acanthaceae, Tokyo, Japan) | Aerial parts | 11 | VSV V-ATPase | VSV MIC 0.66 μM; V-ATPase IC50 0.04–0.49 μM | Asano et al. (1996); Chen et al. (2013) |
Phyllanthus myrtifolius Moon. (Euphorbiaceae, Taiwan, China) | Aerial parts | 12–13 | HIV-1 | 12 EC50 3.5 μM; 13 EC50 5.5 μM | Chang et al. (1995) |
Taiwania cryptomerioides Hayata (Taxodiaceae, Taiwan, China) | aerial parts | 32 | HBV | EC50 1 μM | Yeo et al. (2005) |
Justicia gendarussa Burm. f. and Justicia cf. patentiflora Hemsley (Acanthaceae, Vietnam) | Roots and stems | 151–153 | Drug-resistant HIV-1 | 152, 153 EC50 47–495 nM | Zhang et al. (2017a); Zhang et al. (2017b) |
Symplocos setchuensis Brand. (Sympocaceae, Sichuan, China) | Stems | 154 | HIV replication | EC50 value unavailable | Ishida et al. (2001) |
Parakmeria yunnanensis Hu. (Magnoliaceae, Yunnan, China) | Leaves and stems | 154–156 | HIV | 155 EC50 250 μM 156 EC50 240 μM | Shang et al. (2013b) |
Streblus asper Lour. (Moraceae, Guangxi, China) | Roots | 157, 158 | HBV | 157 anti-HBsAg EC50 3.67 μM; anti-HBeAg EC50 14.67 μM; 158 anti-HBsAg EC50 6.98 μM, anti-HBeAg EC50 26.74 μM | Li et al. (2013) |
Streblus asper Lour. (Moraceae, Guangxi, China) | Stem bark | 158–160 | HBV | 158 HBsAg 6.58 μM, HBeAg 24.86 μM; 159 HBsAg 39.56 μM, HBeAg 61.23 μM | Li et al. (2012a) |
Schisandra propinqua (Wall.) Hook. F. et Thoms. var. sinensis Oliv. (Schisandreae, Sichuan, China) | Aerial parts | 162–165 | anti-HIV-1 | 162 EC50 4.5 μM; 165 EC50 4.5 μM | Li et al. (2012b); Li et al. (2009a); Lei et al. (2007) |
Schisandra sphenanthera Rehd. et Wils (Schisandraceae, Shanxi, China) | Fruits | 166–169 | HSV-2 adenovirus | 166–169 HSV-2 EC50 31.6, 65.7, 65.7, 68.4 μM; adenovirus EC50 59.7, 126.8, 131.3, 100.5 μM | Song et al. (2013) |
Carissa spinarum L. (Apocynaceae, Thailand) | Stems | 170, 249–250, 337, 342, 343, 389 | HSV-1, HSV-2 | EC50 values > 100 μg | Wangteeraprasert et al. (2012) |
Zanthoxylum ailanthoides Sieb. & Zucc. (Rutaceae, Taiwan, China) | Root bark | 171–172 | HIV | EC50 > 238 μM | Cheng et al. (2005b) |
Strobilanthes cusia Bremek. (Acanthaceae, Japan) | Roots | 173–174 | HSV-1 | 173 EC50 > 172 μM; 174 EC50 > 141 μM | Tanaka et al. (2004) |
Polygonum multiflorum Sieb. Et Zucc. (Polygonaceae, Shanghai, China) | Roots | 175, 176 | HIV-1 | 175 EC50 136.1 μM; 176 EC50 162.1 μM | Lin et al. (2010) |
Phyllanthus species (Taiwan, China) | - | 177–179 | HBV | 177 HBsAg and HBeAg EC50 value unavailable, 178 HBsAg EC50 value 36.9 μM, 179 HBsAg EC50 value > 50 μM, 178–179 BeAg EC50 value > 50 μM | Huang et al. (2003a) |
Phyllantthus niruri L. (Euphorbiaceae, Guangxi, China) | Whole plants | 178, 180–181 | HBV | 179–181 HBsAg EC50 97.2, 9.5, 16.7 μM; HBeAg EC50 232.1, 17.4, 69.3 μM | Wei et al. (2012) |
Lindera glauca (Siebold & Zucc.) Blume (Lauraceae, Korea) | Twigs | 171, 224–226 | influenza A/PR8 | 171 and 225 with 30.1% and 29.3% virus infected Vero cells were survived with the treatment of 10 μM | Park et al. (2018) |
Chamaecyparis obtusa (Cupressaceae, Taiwan, China) | Leaves | 227 | HSV-1 | EC50 30.6 μM | Kuo et al. (2006) |
Styrax japonica (Styracaceae, Korea) | Stem bark | 228–230 | HIV-1 virus cell fusion | 228 fusion index 0.25, fusion inhibition 59.4% 229 fusion index 0.22, fusion inhibition 65.1% | Lee et al. (2010a) |
Symplocos setchuensis (Symplocaceae, Sichuan, China) | Stems | 230–232 | HIV | 231 EC50 2.0 μM; 232 EC50 6.9 μM | Ishida et al. (2001) |
Bombax ceiba (Bombacaceae, Guangdong, China) | Roots | 231, 233 | HBV | 231, HBsAg EC50 218.2 μM; 233, HBsAg EC50 123.7 μM | Wang et al. (2013) |
Phenax angustifolius (Urticaceae, USA) | Leaves | 234–238 | HIV | 234–238 EC50 3.0, 5.0, 0.8, 2.8, 5.2 μM | Piccinelli et al. (2005); Rastrelli et al. (2001) |
Phyllanthus acutissima (Euphorbiaceae, Thailand) | Whole plants | 239, 240 | HIV | 239 EC50 27.8 μM; 240 EC50 98.3 μM | Tuchinda et al. (2008) |
Hernandia ovigera (Hernandiaceae, Taiwan, China) | Seeds | 241, 242 | EBV | 470, 480 mol ratio/32 pmol TPA | Ito et al. (2001) |
Phyllanthus species | – | 243, 244 | HBV | inhibition of HBsAg and HBeAg 33.9% and 68.3% at concentration of 50 μM | Huang et al. (2003a) |
Chamaecyparis obtusa (Cupressaceae, Taiwan, China) | Heartwood | 243, 245 | SARS-CoV replication | 243 EC50 > 10 μM, 245 EC50 1.13 μM | Wen et al. (2007) |
Zanthoxylum ailanthoides (Rutaceae, Taiwan, China) | Root bark | 243 | HIV-1 | 243 EC50 < 0.28 μM | Cheng et al. (2005b) |
Arctium lappa L.(Asteraceae, Osaka, Japan) | Fruits | 246, 247 | influenza A virus (A/NWS/33, H1N1) | 246 EC50 24 μM, 247 EC50 3.8 μM | Hayashi et al. (2010) |
Trachelospermum jasminoides (Lindl.) Lem | Stems, leaves | 248 | HCV | EC50 0.87 μM ( HCVcc model), 0.69 μM (HCVpp model) | Qian et al. (2016) |
Brucea javanica (Simaroubaceae, Fujian, China) | Seeds | 250 | TMV | EC50 > 138 μM | Chen et al. (2009) |
Machilus robusta (Lauraceae, Guangxi, China) | Bark | 251–256 | HIV-1 replication | inactive, EC50 value unavailable | Li et al. (2011) |
Schisandra sphenanthera (Schisandraceae, Shaanxi, China) | Fruits | 257–259 | HSV-2, ADV | 257–259 HSV-2, EC50 34.2, 160.1, 35.4 μM; ADV, EC50 68.4, 142.5, 224.7 μM | Song et al. (2013) |
Schisandra sphenanthera (Schisandraceae, Shaanxi, China) | Leaves and stems | 230, 260 | HIV | 233 EC50 31.5 μM | Liang et al. (2013) |
Saururus chinensis rhizomes (Saururaceae, Korea) | Underground part | 261 | HIV-1 | EC50 5.6 μM | Lee et al. (2010b) |
Phyllanthus niruri (Euphorbiaceae, Guangxi, China) | Whole plants | 262 | HBV | HBsAg 15.6 μM, HBeAg 25.1 μM | Liu et al. (2014) |
Schisandra rubriflora (Yunan, China) | Fruits | 281 | HIV-1 | EC50 5.8 μM | Xiao et al. (2010a) |
Kadsura angustifolia (Yunnan, China) | Stems | 259, 282, 283 | HIV-1 | EC50 15.6, 27.0, 21.5 μM | Gao et al. (2008) |
Schisandra propinqua (Sichuan, China) | Aerial parts | 259, 279, 284, 285 | HIV-1 | EC50 4.0, 14.8, 9.4, 2.9 μM | Li et al. (2009b) |
Schisandra chinensis (Heilongjiang, China) | Leaves and stems | 286, 287 | HIV-1 | EC50 > 200 μM | Shi et al. (2014) |
Daphne feddei (Thymelaeaceae, Yunnan, China) | Leaves and stems | 288 | HIV | EC50 23.2 μM | Hu et al. (2011) |
Daphne acutiloba (Thymelaeceae, Yunan, China) | Stems | 289, 290 | HIV | 290 EC50 0.64 μM | Huang et al. (2012) |
Isatis indigotica (Brassicaceae, Anhui, China) | Roots | 291 | influenza, RSV, ADV, PIV3, EV71, HRV | influenza strains after viral adsorption, 137–1137 μM, RSV, ADV, PIV3, EV71, HRV inactive | Yang et al. (2013a) |
Isatis indigotica (Brassicaceae, Anhui, China) | Roots | 292 | influenza A virus A/PR/8/34 (H1N1) | EC50 102 μM | Li et al. (2015) |
Schisandra propinqua (Yunnan, China) | Fruits | 293 | HIV-1 | EC50 15.9 μM | Fan et al. (2010) |
Schisandra propinqua var. sinensis (Sichuan, China) | Aerial parts | 294–297 | HIV-1 | 294 + 295 EC50 7.7 μM | Li et al. (2009b) |
Schisandra propinqua (Hubei, China) | Leaves and stems | 298–306 | HIV-1 IN DNA binding | EC50 > 25 μM | Shang et al. (2013a) |
Schisandra rubriflora (Yunnan, China) | Fruits | 307 | HIV-1 IIIB induced syncytium formationHIV-1IIIB inducted MT-4 cells lytic effects | HIV-1 IIIB induced syncytium formation EC50 30.0 μM HIV-1IIIB inducted MT-4 cells lytic effects EC50 10.8 μM | Xiao et al. (2010a) |
Schisandra chinensis (Heilongjiang, China) | Leaves and stems | 308–309 | HIV-1 | 309 EC50 66.1 μM | Shi et al. (2014) |
Kadsura longipedunculata (Sichuan, China) | Leaves and stems | 310–311 | HIV-1 | 311 EC50 35.9 μM | Pu et al. (2008a) |
Schisandra lancifolia (Yunnan, China) | 312–313 | HIV-1 | 312 EC50 212 μM, 313 EC50 149 μM | Xiao et al. (2010b) | |
Peperomia heyneana (Piperaceae, Yunnan, China) | Whole plants | 314 | HIV-1 | EC50 50 μM | Zhang et al. (2007) |
Saururus chinensis (Saururaceae, Guangxi, China) | Roots | 315–323 | EBV DNA replication | EC50 6.95–69.9 μM | Cui et al. (2014) |
Brucea javanica (Simaroubaceae, Fujian, China) | Seeds | 324–326 | TMV replication | 324–326 EC50 > 100 μM | Chen et al. (2009) |
Parakmeria yunnanensis (Magnoliaceae, Yunnan, China) | Leaves and stems | 327–333 | HIV-1 | EC50 > 80 μg/mL | Shang et al. (2013b) |
Miliusa fragrans (Annonaceae, Thailand) | Leaves and stems | 334–336 | HSV-1, HSV-2 | EC50 > 100 μg/mL | Sawasdee et al. (2013) |
Fraxinus sinboldiana (Oleaceae, Korea) | Stems | 337 | HIV gp41 binding | EC50 > 266 μM | Kim et al. (2002) |
Machilus robusta (Lauraceae, Guangxi, China) | Bark | 326, 338, 339 | HIV-1 | inactive, EC50 value unavailable | Li et al. (2011) |
Phyllanthus virgatus (Taiwan, China) | Whole plants | 340 | HBV | inactive, EC50 value unavailable | Huang et al. (2003a) |
Phyllanthus flexuosus (Euphorbiaceae, Yunnan, China) | Roots | 341 | HSV-1 | inactive, EC50 value unavailable | Zhao et al. (2014) |
Herpetospermum caudigerum (Cucurbitaceae, Tibet, China) | Seeds | 344, 345 | HBV | HBsAg 344 EC50 317 μM, 345 EC50 629 μM, HBeAg 344 EC50 297 μM, 345 EC50 655 μM | Yuan et al. (2005) |
Herpetospermum caudigerum (Sichuan, China) | Seeds | 346–350 | HBV | HBsAg 349 EC50 20.5 μM, 350 EC50 4.89 μM, HBeAg 349 EC50 3.54 μM, 350 EC50 8.02 μM | Yu et al. (2014) |
Vitex leptobotrys (Lamiaceae, Vietnam) | Leaves and twigs | 351, 352 | HIV-1, EBV | 351, 352 anti-HIV-1 inactive; 351 anti-EBV EC50 67 μM | Pan et al. (2014) |
Litsea verticillata (Lauraceae, Vietnam) | Leaves and twigs | 353, 354 | HIV-1, EBV, HCMV | 353 anti-HIV-1 EC50 42.7 μM; anti-EBV EC50 16.2 μM; 354 anti-EBV EC50 22.0 μM; anti-HCMV EC50 58.3 μM | Hoang et al. (2002); Guan et al. (2016) |
Dipsacus asper (Dipsacaceae, Guizhou, China) | Roots | 355–360 | HIV-1 | EC50 > 50 μM | Sun et al. (2015) |
Ligularia kanaitizensis (Asteraceae, Yunnan, China) | Roots and rhizomes | 361, 362 | HIV RT | 362 53.3% inhibition at 439 μM | Li et al. (2005c) |
Hernandia ovigera (Hernandiaceae, Taiwan, China) | Seeds | 363 | EBV | 590 mol ratio/32 pmol TPA | Ito et al. (2001) |
Bombax ceiba (Bombacaceae, Guangdong, China) | Roots | 343, 364 | HBV | HBsAg 343 EC50 18.9 μM; 364 EC50 118.3 μM | Wang et al. (2013) |
Citrus hystrix (Rutaceae, Thailand) | Roots | 365 | HIV-1 | inactive, EC50 value unavailable | Panthong et al. (2013) |
Forsythia suspensa (Oleaceae, Heilongjiang, China) | Fruits | 343, 366–369 | influenza A (H1N1) | inactive, EC50 value unavailable | Li et al. (2014) |
Symplocos setchuensis (Symplocaceae, Sichuan, China) | Stems | 343, 367 | HIV-1 | inactive, EC50 value unavailable | Ishida et al. (2001) |
Calotropis gigantea (Asclepiadaceae, Thailand) | Latex | 367, 370 | influenza A (H1N1) | 370 EC50 24.5 μM | Parhira et al. (2014) |
Rhus javanica (Anacardiaceae, Taiwan, China) | Roots | 343, 365, 367, 371–374 | TMV | 372–374 EC50 218, 280, 193 μM | Ouyang et al. (2007) |
Swietenia macrophylla (Meliaceae, Taiwan, China) | Stems | 375 | HCV | EC50 10.5 μM | Wu et al. (2012) |
Aster flaccidus bge (Asteraceae, Shanxi, China) | Roots | 376 | HIV-1 | EC50 670 μM | Liu et al. (2010) |
Forsythia suspensa (Oleaceae, Heilongjiang, China) | Fruits | 377, 378 | influenza A (H1N1), RSV | influenza A (H1N1), 378 EC50 41.1 μM, RSV 377 EC50 166.4 μM | Li et al. (2014) |
Saururus chinensis (Saururaceae, Korea) | Rhizomes | 379–382 | HIV-1-induced cytopathic effects | 379–381 EC100 1.0, 1.0, 0.2 μM | Lee et al. (2010b) |
Saururus chinensis (Saururaceae, Guangxi, China) | Roots | 321, 379–380, 383–388 | EBV | 321, 379–380, 383–388 EC50 6.95, 3.42, 1.72, 14.5, 7.55, 2.69, 3.52, 1.70, 1.09 μM | Cui et al. (2014) |
Lindera glauca (Siebold & Zucc.) Blume (Lauraceae, Korea) | Twigs | 365, 390–391 | HRV1B and cvb3 | 391 against HRV1B and cvb3 with 75% and 90% virus infected Vero cells were survived with the treatment of 10 μM | Park et al. (2018) |
Parakmeria yunnanensis (Magnoliaceae, Yunnan, China) | Leaves and stems | 392 | HIV-1 | EC50 79.7 μM | Shang et al. (2013b) |
Styrax japonica (Styracaceae, Korea) | Stem bark | 393, 394 | HIV-1 syncytia formation | 393 and 394 percent fusion inhibition of 18.0% and 36.5% at the concentration of 20 μg/mL | Lee et al. (2010a) |
Forsythia suspensa (Oleaceae, Heilongjiang, China) | Fruits | 395–398 | influenza A virus (H1N1) | 396 EC50 60.5 μM | Li et al. (2014) |
Streblus asper (Moraceae, Guangxi, China) | Roots | 399, 400 | HBV | EC50 > 1000 μM | Li et al. (2013) |
Streblus asper (Moraceae, Guangxi, China) | Stem bark | 399 | HBV | EC50 > 1000 μM | Li et al. (2012a) |
Piper regnelli (Piperaceae, Brazil) | Leaves | 392 | BHV-1, poliovirus | EC50 > 170 μM | Bertol et al. (2012) |
Herpetospermum caudigerum (Cucurbitaceae, Tibet, China) | Seeds | 401, 402 | HBV | HBsAg, HBeAg EC50 > 100 μM | Yang et al. (2010a); Yuan et al. (2006) |
Schisandra micrantha (Yunnan, China) | Leaves and stems | 403 | HIV-1 | EC50 9.75 μM | Li et al. (2005a) |
Kadsura angustifolia (Yunnan, China) | Stems | 404, 405 | HIV | EC50 40.86, 13.07 μM | Gao et al. (2008) |
Schisandra lancifolia (Yunnan, China) | Leaves and stems | 406 | HIV-1 | EC50 8.43 μM | Xiao et al. (2010b) |
Schisandra sphenanthera (Shanxi, China) | Leaves and stems | 407 | HIV-1 | inactive, EC50 value unavailable | Liang et al. (2013) |
Illicium henryi (Illiciaceae, Yunnan, China) | Stems and roots | 408–414 | HBV | 409 HBsAg EC50 60 μM | Liu et al. (2011) |
Ailanthus altissima (Simaroubaceae, Fujian, China) | Root bark | 415, 416 | TMV | EC50 > 200 μg/mL | Tan et al. (2012) |
Daphne feddei (Thymelaeaceae, Yunan, China) | Leaves and stems | 417 | HIV-1 | EC50 27.7 μM | Hu et al. (2011) |
Brucea javanica (Simaroubaceae, Fujian, China) | seeds | 416 | TMV | EC50 > 134 μM | Chen et al. (2009) |
Streblus asper (Moraceae, Guangxi, China) | Stem bark | 418–424 | HBV | 424 HBsAg EC50 2.03 μM; HBeAg EC50 3.76 μM | Li et al. (2012a) |
Streblus asper (Sichuan, China) | Heartwood | 423, 425–427 | HBV | EC50 131.23–156.75 μM | Li et al. (2012c) |
Streblus asper (Guangxi, China) | Roots | 428, 429 | HBV | 429 HBsAg EC50 3.14 μM HBeAg EC50 4.74 μM | Chen et al. (2012) |
Streblus asper (Guangxi, China) | Roots | 423, 424, 430–437 | HBV | 423 and 437 EC50 2.03 and 1.58 μM for HBsAg, 3.76 and 3.24 μM for HBeAg, 8.67 and 9.02 μM for DNA replication | Li et al. (2013) |
Kadsura interior (Yunnan, China) | Stems | 442–455 | EBV-EA | 446, 447, 452, 453 showed inhibitory effects on relative ratio 7.1, 2.6, 4.7 and 9.4% at the concentration of 1000 mol ratio/TPA of EBV-EA activation | Chen et al. (2002) |
Kadsura matsudai | Stems | 456–474 | HBV | 456, 457 HBeAg EC50 90.1 and 94.3 μM | Kuo et al. (1999); Li et al. (2000); Kuo et al. (2001); Wu et al. (2003) |
Schizandra arisanensis (Schizandraceous, Taiwan, China) | Stems | 448, 475–477 | HBV | EC50 > 50 μg/mL | Wu et al. (2003) |
Kadsura japonica (Schizandraceous, Taiwan, China) | Stems | 471, 472 | HBV | HBsAg EC50 about 50 μM | Kuo et al. (2005) |
Schisandra rubriflora (Schizandraceous, Yunnan, China) | Fruits | 443, 444, 476–499 | HIV-1 | 478, EC50 11.3; 480, EC50 < 0.65; 481, EC50 2.4; 477, EC50 5.7; 499, EC50 3.9 μM | Chen et al. (2006) |
Schisandra rubriflora (Schizandraceous, Yunnan, China) |
Aerial parts | 443, 444, 477, 480, 488–492, 494, 495, 498 | HIV-1 | 488, EC50 39.4; 489, EC50 36.9; 491, EC50 110 μM | Li et al. (2008) |
Schisandra rubriflora (Schizandraceous, Yunnan, China) | Fruits | 496, 497 | HIV-1 | 496, EC50 4.77; 497 EC50 3.84 μM | Mu et al. (2011) |
Kadsura longipedunculata (Schizandraceous, Yunnan, China) | Roots and stems | 500–504 | HIV-1 | 500 EC50 94.0 μM | Sun et al. (2006) |
Kadsura induta (Schizandraceous, Yunnan Province, China) | Stems | 452, 505–509 | HBV | EC50 > 100 μg/mL | Ma et al. (2007) |
Kadsura angustifolia (Schisandraceae, Yunnan, China) | Stems | 373, 466, 492, 507–509, 577, 510–522 | HIV-1 | 520 EC50 3.86 μM | Gao et al. (2008) |
Kadsura heteroclita (Schisandraceae, Yunnan, China) | Stems | 451, 453, 454, 521, 523–527 | HIV-1 | 451, 527 EC50 3.3, 2.9 μM | Pu et al. (2008b) |
Schisandra sphenanthera Rehd. Et Wils (Schisandraceae, Sichuan, China) | Leaves,stems | 455, 498, 499, 528 | HIV-1 | 455, 498, 499, 528 EC50 35.4, 39.4, 41.5, 35.9 μM | Xiao et al. (2008) |
Schisandra propinqua vas. sinensis (Schisandraceae, Sichuan, China) | Aerial parts | 449, 454, 519, 529–542 | HIV-1 | 449 EC50 9.9 μM; 535 EC50 7.9 μM; 538 EC50 9.1 μM; 540 EC50 7.3 μM; 542 EC50 9.1 μM | Li et al. (2009b) |
Schisandra lancifolia (Schisandraceae, Sichuan, China) | Leaves and stems | 543–545 | HIV-1 | 543–545 EC50 5.40, 9.30, 8.15 μM | Yang et al. (2010b) |
Schisandra wilsoniana (Schisandraceae, Yunnan, China) | Fruits | 455, 477, 482, 546–557 | HBV | 546 inhibitory effects on HBsAg and HBeAg secretion by 59.7% and 34.7% at a concentration of 97 µM | Ma et al. (2009) |
Schisandra wilsoniana (Schisandraceae, Yunnan, China) | Fruits | 553–555 | HIV-1 | 553–555 EC50 8.1, 15.4, 6.9 μM | Yang et al. (2010d) |
Schisandra wilsoniana (Schisandraceae, Yunnan, China) | Fruits | 558–576 | HIV-1 | 558–576 EC50 6.9, 13.6, 13.4, 6.2, 3.6, 3.3, 4.1, 6.5, 6.1, 4.1, 4.7, 3.5, 3.4, 6.2, 5.8, 3.2, 4.2, 3.9 and 4.1 μM | Yang et al. (2010c); Yang et al. (2013b) |
Schisandra wilsoniana (Schisandraceae, Yunnan, China) | Fruits | 443, 444, 447, 455, 476, 482–483, 485, 492, 498, 499, 556, 577–587 | HIV-1, HBV | 447, 498 anti-HIV-1 EC50 3.9, 5.5 μM; 556 anti-HBV on HBsAg and HBeAg secrtion by 59.7 and 34.7% at a concentration of 97 µM | Ma et al. (2013) |
Schisandra neglecta (Schisandraceae, Yunnan, China) | Fruits | 588, 589 | HIV-1 | 588, 589 EC50 4.6, 5.8 μM | Duan et al. (2011) |
Schisandra neglecta (Schisandraceae, Sichuan, China) | Fruits | 583, 585, 590–598 | HIV-1 | 583, 585, 590–598 EC50 4.7, 9.8, 2.2, 1.4, 5.9, 3.5, 8.2, 8.2, 8.3, 11.5, 8.3 μM | Gao et al. (2013) |
Nicotiana tabacum (Solanaceae, Yunnan, China) | Leaves | 599–604 |
HIV-1 TMV |
599–604, anti-HIV-1 EC50 8.8, 4.6, 31.7, 20.4 μM; anti-TMV inhibition rate of 15.2, 58.4, 22.6 and 16.1% at the concentration of 20 μM | Gao et al. (2012) |
Nicotiana tabacum (Solanaceae, Yunnan, China) | Roots,stems | 600–604 | TMV | 600–604 anti-TMV inhibition rate of 14.7, 17.6, 21.4, 22.5 and 23.4% at the concentration of 20 μM | Liao et al. (2012) |
Schisandra wilsoniana (Schisandraceae, Yunnan, China) | Stems | 605–607 | HIV-1 | 605–607 anti-HIV-1IIIB induced syncytia formation EC50 1.5, 4.5, 5.4 μM; 605 reduce p24 EC50 9.0 μM; inhibited primary isolate HIV-1TC-2 replication in PBMCs EC50 1.4 μM | Zhang et al. (2010) |
Schisandra sphenanthera fruits (Schisandraceae, Shaanxi, China) | Fruits | 608–610 | HSV-2, adenovirus | 608–610 anti-HSV-2 EC50 29.1, 122, 35.2 μM, anti-adenovirus EC50 96, 126, 105 μM | Song et al. (2013) |
Schisandra lancifolia (Schisandraceae, Yunnan, China) | Leaves, stems | 611 | HIV-1 | EC50 117 μM | Xiao et al. (2010b) |
Herpetospermum caudigerum (Cucurbitaceae, Sichuan, China) | Seeds | 612 | HBV | HBsAg EC50 0.34 μM, HBeAg EC50 4.83 × 10–4 μM | Yu et al. (2014) |
Parakmeria yunnanensis (Magnoliaceae, Yunnan, China) | Leaves and stems | 613, 614 | HIV-1 | 613 EC50 288 μM | Shang et al. (2013b) |
Peperomia heyneana (Piperaceae, Yunnan, China) | Whole plants | 615–617 | HIV-1 | 615, 616 EC50 5.3, 5.4 μM | Zhang et al. (2007) |
Schisandra sphenanthera (Schisandraceae, Shaanxi, China) | Fruits | 618 | HSV-2, adenovirus | HSV-2, EC50 42.7 μM; adenovirus EC50 55.0 μM | Song et al. (2013) |
Schisandra lancifolia (Schisandraceae, Yunnan, China) | Fruits | 619 | HIV-1 | EC50 1.82 μM | Li et al. (2009a) |
Schisandra lancifolia (Schisandraceae, Yunnan, China) | Leaves and stems | 620 | HIV-1 | EC50 30.3 μM | Xiao et al. (2010b) |
Schisandra lancifolia (Schisandraceae, Yunnan, China) | Leaves and stems | 621, 622 | HIV-1 | 621, 622 EC50 8.6, 7.2 μM | Xue et al. (2011) |
Schisandra wilsoniana (Schisandraceae, Yunnan, China) | Fruits | 623–626 | HIV-1 | 623–626 EC50 10.5, 9.8, 8.9, 7.6 μM | Yang et al. (2013b) |
Kadsura angustifolia (Schisandraceae, Yunnan, China) | Stems | 627, 628 | HIV | 627 EC50 44.68 μM | Gao et al. (2008) |
Daphne feddei (Thymelaeaceae, Yunan, China) | Leaves and stems | 627, 629–630 | HIV | 627, 629–630 EC50 104, 10.0, 8.3 μM | Hu et al. (2011) |
Globoidnan A (2) was considered responsible for the inhibitory activity of HIV integrase of the extract of the buds of Eucalyptus globoidea Blakely (Australia). The lignan was found to inhibit the combined 3′-processing and strand transfer activity of HIV integrase with an EC50 value of 0.64 μM (Ovenden et al. 2004). Diphyllin (11) from Justicia procumbens var. leucantha was determined to have antiviral activity with an MIC value of 0.66 µM against vesicular stomatitis virus (Asano et al. 1996). It was also identified as a novel v-ATPase blocker against influenza viruses (Chen et al. 2013). It could alter cellular susceptibility to influenza viruses through the inhibition of endosomal acidification, thus interfering with downstream viral replication, including that of known drug-resistant strains. Moreover, the combination treatment of the host-targeting diphyllin with pathogen-targeting therapeutics (oseltamivir and amantadine) has been reported to enhance the antiviral effects and cell protection in vitro (Chen et al. 2013; Shen et al. 2011).
Phyllamycin B (12) and retrojusticidin B (13), obtained from Phyllanthus myrtifolius Moon (Euphorbiaceae, Taiwan, China), exhibited inhibitory effects on HIV-1 RT with EC50 values 3.5 and 5.5 µM. They showed less inhibition activities on human DNA polymerase-α (HDNAP-α) with EC50 values more than 200 µM (Chang et al. 1995). Interest in the antiviral activities of 12 and 13 led to the synthesis and bioassay of 18 derivatives (14–31) including a range of nitrogen containing azalignans including 1-aryl-pyrronaphthalenes 23–27 and 3-N-alkylaminomethyl-1-arylnaphthalenes 28–31. The results indicated that compounds 17, 25, 26, and 31 have good activity against HIV, with EC50 values of 0.77, 2.63, 0.02, 0.71 µM, respectively. It was noted that in the 9, 9′-γ-lactone series (16–18), the dicatechol 17 that contains four hydroxyl groups are much more active than the tetramethoxy (16) and dimethylenedioxy (18) derivatives. In addition, the N-isobutyl azalignans 26 and 31 were the most active among the series of compounds 23–31. Interestingly, N-isobutyl-pyrro[4,3-b]naphthalene (26) possessed the highest activity (EC50 = 0.024 µM) with an excellent therapeutic index (TI) (6,000). Therefore, this compound could act as a lead compound for further anti-HIV drug development (Sagar et al. 2004).
Helioxanthin (32) was used as a scaffold to create 45 analogues (33–77) with particular attention paid to the lactone ring and methylenedioxy group, these synthetic analogs were evaluated for their antiviral activities against HBV, HCV, HSV-1, HSV-2, Epstein-Barr virus (EBV), cytomegalovirus (CMV) and HIV (Yeo et al. 2005). Of these analogues, the lactam 49 and the cyclic hydrazide derivative of helioxanthin (59) exhibited significant in vitro anti-HBV activity with EC50 values of 0.08 and 0.03 µM, respectively, and compound 49 was also found to be the most potent HCV inhibitor (55% inhibition at the concentration of 1.0 µM). Compound 43, the acid-hydrolyzed product of the cyclic amide 40, displayed more potent activity than 32 against HBV with an EC50 value of 0.8 µM. In addition, compounds 46 and 53, containing dimethoxy moieties instead of methylenedioxy groups in the C ring of compounds 43 and 49, displayed potent antiviral activities against HCV (64% and 80% inhibition at 3.0 µM, respectively) as well as HBV (EC50 = 0.8 and 0.9 µM, respectively). Compounds 43 and 49 were found to be the strongest HSV inhibitors. They showed inhibitory activity against HSV-1 with EC50 values of 0.15 and 0.29 µM, respectively, and against HSV-2 with EC50 values of < 0.1 and 0.16 µM, respectively. Compound 43 was further determined to have broad-spectrum antiviral activity against HSV-1 (EC50 = 0.15 µM), HSV-2 (EC50 < 0.1 µM), EBV (EC50 = 9.0 µM) and CMV (EC50 = 0.45 µM). This compound was approximately 140 and 210 times more potent than the reference drug acylovir (EC50 = 21 µM) against HSV-1 and HSV-2, respectively. The cyclic hydrazide 59 and its brominated product 73 showed anti-HIV activities with EC50 values of 2.7 and 2.5 µM, respectively (Yeo et al. 2005). Moreover, as established by quantification of viral DNA, RNA, covalently closed circular DNA, and protein synthesis, compound 59 inhibited duck HBV (DHBV) activity without affecting the stability of cellular macromolecules, and it had a sustained antiviral effect even after drug removal. When DHBV replication was induced, virus-harbouring cells were more susceptible to the cytotoxicity of the compound than the non-induced cells (Ying et al. 2010).
In a study of inhibitors of HBV gene expression and replication, compound 32 and its lactam analogue 49 demonstrated strong anti-HBV activity by markedly decreasing HBV DNA, 3.5-kb and 2.4/2.1-kb RNA, and the core protein in HepG2.2.15 cells. Compounds 32 and 49 revealed EC50 values of 1.0 and 0.08 µM against HBV DNA, and 1.0 and 0.09 µM against HBV RNA (3.5 kb), respectively. In addition, western blot analysis of the cell lysate from HepG2.2.15 cells confirmed that the core protein expression decreased in a dose-dependent manner after drug treatment. Moreover, the EC50 values for compounds 32 and 49 as inhibitors of HBV DNA production were determined to be 0.4 µM and 0.004 µM against the drug-resistant mutant W10, and 0.1 µM and 0.0003 µM against the drug-resistant mutant DM2, respectively (Li et al. 2005b; Cheng et al. 2005a; Tseng et al. 2008).
In a study of the anti-HBV mechanism, compound 59 was found to suppress HBV RNA and protein expression as well as DNA replication of both the wild-type and 3TC-resistant virus. In addition, the time-course analyses revealed that RNA expression was inhibited first after treatment with compound 59, followed by viral proteins, and then DNA. This compound inhibited the activity of all HBV promoters by decreasing the binding of hepatocyte nuclear factor (HNF)-4, HNF-3, and fetoprotein factor to the precore/core promoter enhancer II region. Therefore, compound 59 suppressed HBV replication by posttranscriptional downregulation of critical transcription factors in HBV-producing cells, thus reducing HBV promoter activity and blocking viral gene expression and replication (Ying et al. 2007; Quasdorff & Protzer 2010).
Yeh et al. also synthesized series of helioxanthin analogues (78–110) by modification at the lactone rings. These compounds were evaluated for their anti-HBV activity in HepA2. Among them, helioxanthin (32) and the derivative 103 were found to possess the best activity (EC50 = 0.16 µM and 0.14 µM, respectively), whereas the retro-isomer 79 (EC50 = 2.18 µM) is less active. Interestingly, compound 105 differs from 103 only by switching the locations of the hydroxyl (OH) and the methoxy (OCH3) groups, yet its anti-HBV activity was markedly reduced to an EC50 value of 1.89 µM. The γ-lactam ring opening analogues 84, 89, 90 and 91 exhibited antiviral activity against HBV with EC50 values of 0.89, 1.41, 0.42 and 1.22 µM, respectively. Analogues 93, 108 and 109 exhibited moderate to low anti-HBV effects (EC50 = 2.39, 3.06 and 7.64 µM, respectively), while all other compounds did not display detectable anti-HBV effects within the same range of concentrations. Compound 103 potently suppressed both endogenously expressed HBsAg and HBeAg production with EC50 values of 0.06 and 0.14 μM, respectively. It not only inhibited HBV DNA with wild-type (stably transfected with 1.3-fold wild-type HBV ayw strain genome in HepG2 cells) and lamivudine-resistant (HepG2 cell line [HBV] containing the lamivudine-resistant HBV with L515M/M539V double mutation in the DNA polymerase region) strains, but also suppressed all HBV transcripts and HBV core protein in 1.3 ES2 cells. Additionally, it could selectively suppress viral promoter activity for HBV surface antigen, core antigen and enhancer I in HepA2 cells (Janmanchi et al. 2010).
Helioxanthin analogues 103, 105, 108, 111–150) were also synthesized and evaluated for their anti-hepatitis B virus activity. Among them, 112 exhibited the strongest inhibition (EC50 value 0.06 μM), which was about three times more active than its parent compound (32). Potent activities were also found for the fluorine containing 117, the γ-lactone ring opening 129–131 and the lactone group containing 147 with EC50 values of 0.38, 0.67, 0.49, 0.65 and 0.86 μM, respectively. From these synthesized arylnaphthalene lignans, no distinguished structure–activity relationship could be concluded. However, it is obvious that the retro-lactonization and the ring opening of the lactone group do not lower the anti-HBV activity. The most effective compound, 112, not only suppressed HBsAg production in HepA2 cells and viral replication of HBV (stably transfected with 1.3-fold wild-type HBV ayw strain genome in HepG2 cells), but also inhibited all HBV transcripts in 1.3ES2 cells and viral core promoter activity in HepA2 cells (Janmanchi et al. 2013).
Justiprocumins A (151) and B (152), patentiflorin A (153) were obtained from Justicia gendarussa Burm. f. and Justicia cf. patentiflora Hemsley (Vietnam). Compounds 152 and 153 displayed potent activity against a broad spectrum of HIV strains with EC50 values in the ranges of 15–37 nM (AZT, EC50 77–95 nM). They also showed potent inhibitory effects against drug-resistant HIV-1 isolates (HIV-11617–1/HIV-1N119) of both the nucleotide analogue (AZT) and non-nucleotide analogue (nevaripine) with EC50 values in the ranges of 47–495 nM (Zhang et al. 2017a, 2017b). In this study, the C-7 sugar unit-containing compounds (151, 153) display better anti-HIV activity than their aglycone diphylin (11). Compound 153 also exhibited potent inhibitory activity against Zika virus (ZIKV) infection in vitro and in vivo, a mosquito-borne flavivirus that causes microcephaly and severe brain malformations in fetus. It blocked ZIKV infection in African greenmonkey kidney epithelial cells (VERO), human fibroblast cells (HT1080), and human microglial cells (CHME3) with EC50 values between 0.01–0.03 μM. The result was confirmed in vivo by using type I interferon receptor knockout mouse model C57BL/6 Ifnar1−/−. Compound 153 also showed a broad spectrum inhibition against other Flaviviridae viruses including DENV1, tick-borne encephalitis virus (TBEV), West Nile virus (WNV), JEV and EBV with the EC50 values ranged between 0.12–1.0 μM (Martinez-Lopez et al. 2019).
Aryltetralin-type lignans
Aryltetralin lignans have a 10-membered bicyclic core, with two aromatic ring systems. Unlike arylnaphthalene lignans, which involve all three rings being aromatized, aryltetralin lignans have a variety of configurations possible in the non-aromatic ring (ring B) formed by fusion of the two phenylpropanoid units. This leads to a greater diversity of possible isomers and stereochemical outcomes in aryltetralin lignans. We have included lignans bearing a dialin ring-system in this section, since they also contain stereocenters in the core of the molecule and are not typically recognized as a separate class of lignans. The most well-known lignan of this type of compounds may be podophyllotoxin. The compound was discovered from Podophyllum peltatum L. in 1880 (Cragg et al. 2011). Two of its derivatives, etopside and teniposide, were granted as anticancer medications by the US FDA in 1983 and 1992, respectively (Hande 1998; Cragg and Newman 2005).
During the time covered by this review, 31 aryltetralin lignans isolated from plants were tested for antiviral activity (154–181, 224–226) (see Table 1 for source plants, antiviral activities, and references). Of the plant-derived aryltetralin lignans, only compounds 157, 162, and 165 displayed antiviral activity at concentrations below 5 μM. There were three studies on synthetic podophyllotoxin analogues which displayed significant antiviral activities and provided information about structure–activity relationships.
In an ongoing study of the chemical composition of Streblus asper Lour. (Moraceae, Guangxi, China), Li et al. isolated (-)-isolariciresinol (157) and 9-β-xylopyranosyl-(-)-isolariciresinol (158) from the plant’s roots. In tests for potential inhibition of the secretion of HBV s antigen (HBsAg) and HBV e antigen (HBeAg), and the replication of HBV DNA in HBV-infected HepG2.2.15 cells, compounds 157 and 158 displayed anti-HBV activity of EC50 values of 3.67 and 6.98 μM for HBsAg with TI values 35.28 and 30.15, and EC50 values of 14.67 and 26.74 μM for HBeAg with TI values 8.82 and 7.87, respectively. In addition, the compounds showed inhibitory activity on the replication of HBV DNA with EC50 values of 12.72 and 17.68 μM and TI of 10.18 and 11.90, respectively (Li et al. 2013). Compounds 158, 5-methoxy-9-β-xylopyranosyl-(-)-isolariciresinol (159) and 9-β-xylopyranosyl-lyoniresinol (160) were obtained from the stem bark. The only difference of 160 from 158 is that it contains an extra methoxyl group in ring C, and its difference from 159 is that it is lacking an methoxyl group in ring A. Compounds 158 and 159 inhibited the expression of HBsAg and HBeAg with EC50 values of 6.58 and 39.56 μM for HBsAg (TI > 2), and 24.86 μM (TI = 8.06) and 61.23 μM (TI = 1.75) for HBeAg, respectively, while 160 showed no activity against HBV at the concentration of 1000 μM (Li et al. 2012a).
Three new aryltetralin lignans, sinensisins A-C (162–164) and the known (8R,7′R,8R)-5-hydroxy-4,3′,4′-trimethoxy-2,7′-cyclolignan (165) were isolated from the aerial parts of Schisandra propinqua (Wall.) Hook. F. et Thoms. var. sinensis Oliv. (Sichuan, China) and tested for HIV-1 inhibitory effects in a microtiter syncytium formation infectivity assay. Among them, both 162 and 165 displayed anti-HIV-1 activity with EC50 values of 4.5 μM and TI values of 6.7 (Li et al. 2012b, 2009a; Lei et al. 2007).
Thirteen podophyllotoxin derivatives (182–194) containing modifications in the phenyl C-ring or the lactone ring were prepared and evaluated for their antiherpetic activity against HSV-2 on Vero cells. Among them, phenazines 192 and 193 showed the highest antiviral potency against HSV-2 (Castro et al. 2003).
A series of derivatives (195–216) of podophyllotoxin with structural modifications mostly at C-7 were synthesized and tested for their inhibitory effects of HIV-1 replication in acutely infected H9 cells. Among the derivatives, the 4-amino derivatives 213–216 were the strongest HIV inhibitors exhibiting EC50 values of < 0.002 μM and TI values > 120. The 4-amino substituted compounds 199 and 203 also displayed strong HIV activity with EC50 values of 0.021 and < 0.002 μM and TI values of 19.1 and > 16, respectively. The 4β-hydroxyl containing compound 197 and its corresponding vinyl carbamate 208 were the strongest HIV inhibitors among the non-nitrogen containing derivatives with EC50 values of 0.14 and 0.072 μM, respectively. However, compound 210, which contains a 4α-hydroxyl group showed less inhibitory activity with an EC50 value of 0.90 μM and a TI value of 19.4. Comparison of the anti-HIV activity of these derivatives suggested that the methylenedioxy group of C-4 and C-5 being substituted by two methoxy groups enhanced the anti-HIV activity (Zhu et al. 2004).
In 2007, a new series of podophyllotoxin derivatives (217–223) some of which contained the antiviral drug molecule stavudine and different structural podophyllotoxin analogues were designed, synthesized, and tested for their inhibitory activity against HIV-1 replication in acutely infected C8166 cells by Tu et al. Among these compounds, 219 and 220 showed the highest anti-HIV-1 activities with EC50 values of 0.29 and 0.17 μM and TI values of 354.5 and 466.9, respectively. Moreover, the 7β–cyano-substituted 4,5–dimethoxy compound 222 also showed promising anti-HIV activity (EC50 = 1.05 μM and TI = 131.28). However, compound 223, which is a carboxylic acid derivative of podophyllotoxin obtained by hydrolyzation of 222, was significantly less active than 222 with an EC50 value of 46.90 μM and a TI value > 9.31 (Chen et al. 2007).
Dibenzylbutyrolactone-type lignans
Dibenzylbutyrolactone lignans are defined by the presence of a five-membered lactone ring formed by the cyclization of oxidized C-9 and C-9′ carbons. Similar to the aryltetralin lignans, this type of lignan may possess a wide variety of relative and absolute configurations, leading to a large number of isomers. The lactone ring in dibenzylbutyrolactone lignans is reminiscent of the lactones frequently observed in the structures of the previously discussed arylnaphthalene and aryltetralin lignans. Most of this type of lignan showed insignificant antiviral activity, although a few of them were studied in mice for their antiinfluenza activity.
From 1998 to 2019, 22 dibenzylbutyrolactone lignans were isolated from plants and tested for antiviral activity (227–248) (see Table 1 for source plants, antiviral activities, and references). Compounds 236, 243, 245, and 248 are worthy of additional discussion due to their potent antiviral activity (≤ 1 μM). Interestingly, considering the potent activity of the four aforementioned potently active compounds, we could find no studies on antiviral activities of synthetic dibenzylbutyrolactone lignans.
Phenaxolactones 1–5 (234–238) were isolated from the leaves of Phenax spp. (Urticaceae, USA). Compound 234 showed anti-HIV activity with an EC50 value of 3.0 µM and TI value of 37.3. Compound 236 exhibited increased anti-HIV activity with an EC50 value of 0.8 µM but lower TI value of 12.5, while 237, the glycosylated derivative of 236, showed less activity with an EC50 of 2.8 µM and TI of 6.4. Moderate antiviral activity was observed for 235 (EC50 of 5.0 µM and TI of 15.0) and 238 (EC50 of 5.2 µM and TI of 14.4) (Piccinelli et al. 2005; Rastrelli et al. 2001).
Hinokinin (243), isolated from a Phyllanthus species, significantly inhibited both HBsAg and HBeAg production with inhibitions of 33.9% and 68.3% at a concentration of 50 µM, respectively. ( +)–trans–8-(3,4-(Methylenedioxybenzyl)-8′-(3′,4′-dimethoxybenzyl)-butyrolactone (244), and a tannin virgatyne were found to have no antiviral activity (Huang et al. 2003a). Purified from the heartwood of Chamaecyparis obtusa (Cupressaceae, Taiwan, China), 243 and savinin [245, the enantiomer of isogadian (240)] showed high inhibitory activity in the cell-based assay utilizing CPE on Vero E6 cells via SARS-CoV infection at the concentration of 20 μM. In the evaluation for inhibition of SARS-CoV replication using ELISA, 243 had an EC50 value higher than 10 μM, but the EC50 value of 245 was determined to be much more potent with a value of 1.13 μM (TI > 667). In the test for inhibitory effect of SARS-CoV 3CL protease activity, 243 and 245 displayed EC50 values of > 100 and 25 µM, respectively, and a Ki value for 245 was determined to be 9.1 μM in the study of the inhibitory mechanism (Wen et al. 2007). Isolated from the root bark of Zanthoxylum ailanthoides (Rutaceae, Taiwan, China) as well, hinokinin (243) displayed significant anti-HIV-1 activity with an EC50 value of < 0.28 μM and a TI value of > 18.7 in the test for inhibition of HIV replication (Cheng et al. 2005b).
Trachelogenin (248), obtained from the stems and leaves of Trachelospermum jasminoides (Lindl.) Lem., exhibited anti-HCV potential with EC50 values of 0.87 μM (in HCVcc model) and 0.69 μM (in HCVpp model). Compound 248 was a HCV entry inhibitor by interfering with the interactions between HCV glycoprotein E2 and the host entry factor CD81 (Qian et al. 2016).
Dibenzylbutane-type lignans
Dibenzylbutane lignans bear the least modified connections between the 8 and 8′ carbons of the phenylpropanoid precursors. This class also contains a large number of stereoisomers due to the chiral carbons created by linking C-8 to C-8′. The carbons derived from C-9 and C-9′ are found in a variety of oxidation states varying from methyl groups to carboxylic acid functionalities. As with benzylbutyrolactones, this type of lignans is also lacking lead molecules that show attractive antiviral activities.
We found records of 29 dibenzylbutyrolactone lignans isolated from plants and tested for antiviral activity (249–262, 269, 280–287) (see Table 1 for source plants, antiviral activities, and references). Compounds 259 and 285 displayed the most activity in antiviral assays (< 5 μM). There were 10 synthetic dibenzylbutryolactones published between 1998 and 2020, most of which were modeled off of nordihydroguaiaretic acid (NDGA) (269).
Nordihydroguaiaretic acid (NDGA) (269), a lignan obtained from the leaves and twigs of Larrea tridentate, was evaluated for its anti-dengue virus (DENV) effects. It displayed a dose-dependent inhibitory effects on the viral yield and nonstructural protein-1 secretion in the supernatants of infected cells treated for 24 and 48 h. In DENV4 replicon-transfected Vero cells, treatment with 269 at 50 and 100 μM significantly reduced DENV replication. Furthermore, treatment with this compound not only led to the reduction in the number of lipid droplets (LDs), the neutral lipid storage organelles involved in DENV morphogenesis that are known to proliferate during DENV infection, but also resulted in dissociation of the C protein from LDs (Soto-Acosta et al. 2014). Compound 269 was assessed for its ability to elicit HIV replication. It was demonstrated to increase the HIV-1 p24 antigen two fold at the concentration of 15 μM. In addition, the treatment of 269 at 15 μM also increased reactive oxygen species production in U1 cells up to 140% compared with control cells (Barquero et al. 2014). Compound 269 was tested as an HPV16 E6 gene inhibitor. This compound was shown to inhibit HPV16 E6 mRNA expression, influence E6 gene transcription, and induce decreased protein expression levels of E6 and p53 (Chen et al. 2008). In an investigation on host lipid/fatty acid synthesis and the HCV life cycle, 269 canceled the HCV-induced alteration of host lipid homeostasis. This compound not only reduced sterol regulatory element-binding protein activation and increased the expression of the genes involved in β-oxidation, but it also inhibited very-low-density lipoprotein (VLDL) secretion by affecting mediators of VLDL biosynthesis. Whereas HCV induced the accumulation and perinuclear distribution of LDs, 269 decreased the overall number and increased the average size of the LDs (Syed and Siddiqui 2011). Furthermore, when 269 was added simultaneously with the virus, it could inhibit Junin viral replication (Konigheim et al. 2005).
Tetra-O-glycyl-NDGA (270), a water-soluble derivative of 269, competed effectively with the DNA binding domain of recombinant Sp1 protein to bind to the HIV long terminal repeat, as determined by an electrophoretic mobility-shift assay. By inhibiting Sp1 binding to the HIV long terminal repeat, 270 repressed Sp1-regulated HIV Tat transactivation and replication in cultured cells, with an EC50 of 12 μM. Moreover, replication of simian immunodeficiency virus was completely blocked by 270 at a concentration of 5.0 μM. Furthermore, at 100 μM, 270 was harmless towards 174 × CEM cells and H9 cells (Huang et al. 2003b).
Nine synthesised NDGA derivatives 271–277, Mal.4 (278), and M4N (279) inhibited Tat transactivation in a dose-dependent manner. At 80 μM, these compounds blocked Tat-regulated secreted alkaline phosphatase production by > 90%; and at 20 μM, the inhibition was still > 50%. All of these newly synthesized derivatives showed a greater inhibitory activity than the parent NDGA 269 (EC50 = 20 μM) and Mal.4 (278, EC50 = 25 μM). Except for compounds 271 (EC50 = 17.2 μM) and 273 (EC50 = 17.3 μM), all other NDGA derivatives demonstrated greater potency than M4N (279, EC50 = 11.1 μM). Compound 272 (EC50 = 0.88 μM) was determined to be the strongest inhibitor of HIV Tat-regulated transactivation among all of the new NDGA derivatives (Hwu et al. 2008). Furthermore, three synthesized NDGA derivatives, tetra-acetyl NDGA (280), 278 and 279, were all demonstrated to inhibit gene expression from the early promoter P97 of HPV16. Using luciferase activity as the indicator of gene expression, compounds 278 and 279 were shown to be active in a dose-dependent manner. However, 280 (EC50 = 11 μM) was shown to be a better inhibitor of the HPV P97 promoter activity than 278 (EC50 = 37 μM) and 279 (EC50 = 28 μM). These compounds demonstrated very little effect on the gene expression guided by the ADV major late promoter and the CMV promoter (Craigo et al. 2000; Halim et al. 2013). Compound 279 inhibited 10 passages of HSV-1 and 4 passages of HSV-2 with the EC50 values ranges 4–11.7 μM, while the EC50 ranges of ACV increased from 7 μM to 444 μM. The results indicated that 279 had less drug resistance than ACV. Compound 279 inhibited HSV by decreasing the expression of the HSV immediate early gene α-ICP4 with an EC50 value of 43.5 μM, which was essential for HSV replication (Chen et al. 1998).
Compound 281, 3,3′-Dihydroxy-4,4′,5,5′-tetramethoxylignan, was isolated from the fruits of Schisandra rubriflora (Yunan, China). It showed inhibitory activity on HIV-1IIIB induced syncytium formation with an EC50 value of 5.8 µM and a TI value of 4.46 (Xiao et al. 2010a). Kadangustins H (282) and I (283), meso-dihydroguaiaretic acid (259), tiegusanin N (284), meso-monomethyldihydroguaiareticacid (285) and 279 were isolated from the stems of Kadsura angustifolia (Yunnan, China). The compounds showed anti-HIV activity with EC50 values of 27.0, 21.5, 15.6/4.0, 2.9 and 14.8 μM, and TI values of 2.8, 1.1, 3.2/6.2, 5.8 and 2.0, respectively (Gao et al. 2008; Li et al. 2009b).
Tetrahydrofuranoid and tetrahydrofurofuranoid-lignans
Tetrahydrofuranoid lignans have a characteristic five-membered oxygen-containing ring connecting the two phenylpropanoid precursor units. This ring can be derived from a C-7 and C-8 to C-7′ and C-8′ linkage (7,8,8′,7′-tetrahydrofuranoid lignans, compounds 294–311, 313–323, 334–336, 338, 340, 379–388), a C-7 and C-8 to C-8′ and C-9′ linkage (7,8,8′,9′-tetrahydrofuranoid lignans, compounds 291, 292, 324, 325, 327, 337, 339, 344, 346, 347, 376–378, 390 and 391), or a C-8 and C-9 to C-8′ and C-9′ linkage (9,8,8′,9′-tetrahydrofuranoid lignans, compounds 288–290, 333 and 389). This diversity of precursor orientations, combined with the possibility of stereoisomers leads to a large diversity of possible isomers being isolated. Tetrahydrofurofuranoid lignans are defined by the presence of a bicyclic furofuran ring-system formed by the connection of C-7 to C-9′ through an oxygen atom, and a connection between C-9 and C-7′ through an oxygen atom (tetrahydrofurofuranoid lignans, compounds 293, 312, 326, 328–332, 341–343, 345, 348–375).
The antiviral activity of 104 plant-derived tetrahydrofuranoid and tetrahydrofurofuranoid lignans were reported between 1998 and 2020 (288–391) (see Table 1 for source plants, antiviral activities, and references). Five compounds in this class (290, 379–381, 388) displayed antiviral activity at concentrations of 1 μM or less. We did not find any antiviral studies on synthetic tetrahydrofuranoid or tetrahydrofurofuranoid lignans during the time period covered in this review.
4,4′-Dihydroxy-3,3′-dimethoxy-9-ethoxy-9,9′-epoxylignan (289) and daphnenin (290), are two lignans that were isolated from the stems of Daphne acutiloba (Thymelaeceae, Yunan, China). Compound 290 showed anti-HIV activity with an EC50 value of 0.64 µM (Huang et al. 2012).
Manassantins A–B (379–380) and saucerneols B–C (381–382) were isolated from Saururus chinensis (Saururaceae, Korea) rhizomes and evaluated for their anti-HIV-1 activities. Compound 379 showed dose-dependent inhibitory activities on HIV-1 protease with an EC50 value of 38.9 μM. Compounds 379–381 inhibited HIV-1-induced cytopathic effects in a human T lymphoblastoid cell line with EC100 values of 1.0, 1.0 and 0.2 μM, respectively. Compound 381 showed the most potent and selective anti-HIV-1 activity with an EC100 value of 0.2 μM, a CC0 value of > 125.0 μM, and a TI value of > 520.8 (Lee et al. 2010b). Nine tetrahydrofuran lignans, (–)–(7"R,8"R)–saucerneol J (321), manassantins A–B (379–380), saurucinol B (383), 4′′-O-demethylmanassantin A (384), 3′′-O-demethylmanassantin B (385), 4–O–demethylmanassantin B (386), saucerneol methyl ether (387) and saucerneol D (388) were isolated from the roots of S. chinensis (Saururaceae, Guangxi, China). In the examination for their abilities to inhibit EBV lytic DNA replication in P3HR-1 cells, these compounds showed strong to moderate activities with EC50 values of 6.95, 3.42, 1.72, 14.5, 7.55, 2.69, 3.52, 1.70 and 1.09 μM, and TI values of 3.3, 58.5, 116.4, 4.4, 9.2, 20.2, 20.2, 65.0 and 41.1, respectively (Cui et al. 2014).
Benzofuran lignans
Benzofuran lignans are determined by the presence of a furan juncture created from the linkage of C-7 to C-4′ through an oxygen atom and the direct linkage of C-8–C-3′. Due to the phenylpropanoid precursor units being connected at carbons other than C-8 to C-8′, these compounds are sometimes classified as 8,3′–neolignans. In addition to the isomeric possibilities caused by the stereogenic center in the 7, 8–dihydrofuran ring, there are also many possible modifications of the propyl chain of this class of molecules.
There were 26 benzofuran-type lignans isolated from plants and tested for antiviral activity (392–417) in the time period covered by this review (see Table 1 for source plants, antiviral activities, and references). The benzofuran lignans tested between 1998 and 2020 displayed the least antiviral activity of all of the classes of lignans. The most active benzofuran lignans were 403 and 406, however, there were none with EC50 values ≤ 5 μM. There were no antiviral studies on synthetic lignans of this class.
Vladinol F (403), a lignan isolated from the leaves and stems of Schisandra micrantha (Yunnan, China), displayed anti-HIV-1 activity with an EC50 value of 9.75 μM and a TI value of 27.45 (Li et al. 2005a). Balaphonin (406) was obtained from the leaves and stems of S. lancifolia (Yunnan, China). It showed anti-HIV-1 effects with an EC50 value of 8.43 μM and a TI value of 4.3, respectively (Xiao et al. 2010b).
Neolignans
Neolignans are phenylpropanoid dimers connected at positions other than C-8/C-8′. Here, we are focusing on neolignans biosynthesized by connection of the C-3/C-3′ carbons in the phenylpropanoid precursors, or via an ether linkage.
Compounds 418–436 represent the 19 neolignans isolated from plants and tested for antiviral activity between 1998 and 2020 (see Table 1 for source plants, antiviral activities, and references). There were three plant-derived neolignans that exhibited EC50 values ≤ 5 μM (424, 429, 436). Among this type of compounds, 424 may be considered as a lead molecule against HBV infection with a high TI value. There were 5 synthetic neolignans (437–441) made and assayed for antiviral activity.
Seven neolignans, (7′S,8′S)–trans–streblusol A (418), (7′R,8′S)–erythro–streblusol B (419), (7′S,8′S)–threo–streblusol B (420), 8′R-streblusol C (421), (8R,8′R)-streblusol D (422), threo-strebluslignanol (423) and magnolol (424) were isolated from the stem bark of Streblus asper (Moraceae, Guangxi, China). Magnolol (424) showed anti-HBV activities against HBsAg antigen with an EC50 value of 2.03 μM and a TI value of 31.37, and HBeAg antigen with an EC50 value of 3.76 μM and a TI value of 316.94 (Li et al. 2012a). (7R,8S,7′R,8′S)–erythro–Strebluslignanol H (428), -and honokiol (429) were isolated from the roots of the same plant. Among them, 429 exhibited anti-HBV activities with EC50 values of 3.14 μM (TI = 21.5) on HBsAg and 4.74 μM (TI = 14.2) against HBeAg (Chen et al. 2012). Compound 429 also displayed an antiviral effect against HCVcc infection at non-toxic concentrations. It inhibited the cell entry of lentiviral particles pseudo-typed with glycoproteins from HCV genotypes 1a, 1b, and 2a, but not the VSV. The expression levels of the components of replication complex, NS3, NS5A and NS5B, were down-regulated by 429 in a dose-dependent manner in the concentration range of 10–30 μM. The compound inhibited HCV replication dose dependently in both genotypes 1b and 2a sub-genomic replicons in the concentration of 10 and 20 μM. Therefore, it was determined that 429 inhibited HCV infection by targeting cell entry and replication with an EC50 value of 1.2 μM, an EC90 value of 6.5 μM, and a TI (LD50/EC90) value of 5.4 (Lan et al. 2012). Furthermore, during the evaluation for activity against anti-SARS-CoV using a cell-based assay measuring SARS-CoV-induced cytopathogenic effects on Vero E6 cells, 424 and 429 showed inhibitory activity at the concentrations between 10 and 20 μM by cell-based CPE assay, and they displayed appreciable levels of anti-SARS virus bioactivity with EC50 values ranging from 3.8 to 7.5 μM and the CC50 values of > 65 μM (Wen et al. 2007).
In a separate study, 12 neolignans including 423, 424, magnolignan A (430), magnolignan A-2-O-β-D-glucopyranoside (431), isostrebluslignanaldehyde (432), isomagnolol (433), obovatol (434), strebluslignanol F (435) and (7′R,8′S,7″R,8″S)-erythro-strebluslignanol G (436) were isolated from the roots of S. asper (Guangxi, China) and evaluated for their anti-HBV activities using the HBV transfected HepG2.2.15 cells. The neolignan 424 and the dimeric neolignan 436 displayed activities against anti-HBV with EC50 values of 2.03 (TI = 31.37) and 1.58 μM (TI = 74.90) for HBsAg, and with EC50 values of 3.76 (TI = 16.94) and 3.24 μM (TI = 36.52) for HBeAg. They also showed HBV DNA replication with EC50 values of 8.67 (TI = 7.34) and 9.02 μM (TI = 13.12), respectively (Li et al. 2013).
Five 1,4–benzodioxane neolignans eusiderin A (437), eusiderin B (438), eusiderin G (39), eusiderin M (440) and nitidanin (441) were synthesized and screened for their anti-HCV activity. They displayed inhibitory effects with the EC50 values of 30, 20, 25, 50 and 200 μM and the TI values of 1.21, > 6.25, 1.91, 2.57 and 2.32, respectively (Pilkington et al. 2018).
Dibenzocyclooctadiene lignans and homolignans
Dibenzocyclooctadiene lignans are defined by having a C-2/C-2′ linkage as well as the usual C-8/C-8′ juncture of the phenylpropanoid precursor units. This endows them with a cyclooctadiene ring. Unlike dibenzocyclooctadiene lignans, which have both aromatic rings intact, homolignans lack one of the 6-carbon aromatic ring moieties. In some cases, homolignans possess a cyclohexadienone moiety, and in some instances, the cyclohexadienone moiety has been cleaved.
Between 1998 and 2020, 157 dibenzyocyclooctadiene lignans and homolignans (442–598) were isolated from plants and tested for antiviral activity, by far the most of any class of lignan over this time period (see Table 1 for source plants, antiviral activities, and references). This class of lignan had 20 compounds with antiviral activity (≤ 5 μM), but the most interesting, due to their potent activities (EC50 ≤ 1 μM), are 480 and 591. There were no studies on synthetic dibenzyocyclooctadiene lignans or homolignans.
The lignans interiotherin B (443), angeloylgomisin R (444), schisantherin B (476), ( +)-gomisin K3 (477), rubrisandrins A and B (478–479), ( ±)-gomisin M1 (480), ( +)-gomisin M2 (481), deoxyschisandrin (482), schisandrin (483), tigloylgomisin P (484), gomisin O (485), angeloylgomisin P (486), epigomisin O (487), methylgomisin R (488), ( +)-14-tigloylgomisin K3 (489), 12-demethylwuweilignan I (490), schisandrene A (491), gomisin R (492), (R)-( +)-gomisin M1 (493), ( +)-angeloylgomisin K3 (494), dimethylgomisin J (495), rubrilignans A-B (496–497), schisantherin D (498), and gomisin J (499) were obtained from the extract of Schisandra rubriflora and their anti-HIV effects were evaluated (Chen et al. 2006; Li et al. 2008; Mu et al. 2011). Among them, rubrisandrin A (478), gomisin J (499), ( ±)-gomisin M1 (480), ( +)-gomisin M2 (481) and ( +)-gomisin K3 (477) were active in the HIV growth inhibition assay with EC50 values 11.3, 3.9, < 0.65, 2.4 and 5.7 μM, and TI values of > 5.7, 6.0, > 68, 19.4 and 7.4, respectively (Chen et al. 2006). Compound 480 possessed anti-HIV activity against a wide variety of HIV-1 and HIV-2 viral strains such as NL 4–3 (X4), Bal (R5), (B) BK132 (X4), (AE) 92TH001 (R5) and CBL-23 with EC50 values in the range of 1–3 µM. Further investigation indicated that 480 was a non-nucleoside reverse transcriptase inhibitor (NNRTI). The studies in TZM-bl indicator cells showed 480 exerted inhibitory activity against both NL 4–3 and BaL, suggesting that it targets an early step in the HIV life cycle. Quantitative real-time PCR demonstrated that 480 blocked both early and late HIV-1 reverse transcription products (Han et al. 2015). Compounds 496 and 497 showed anti-HIV-1 activities with EC50 values of 4.77 and 3.84 µM, and TI of 35.5 and 18.6, respectively (Mu et al. 2011).
Phytochemical investigation of Kadsura angustifolia (Schisandraceae, Yunnan, China) led to 19 dibenzocyclooctadiene lignans kadangustins A-G (510–516), schisantherins Q (517) and L (507), gomisin R (492), deangeloylschisantherin F (518), schisantherin F (519), binankadsurin A (520), kadsulignan K (521), epoxideschisandrin C (522), kadsurarin (373), kadsulignan N (466), schisantherin P (508) and kadsulignan L (509). Compound 520 showed anti-HIV activity with an EC50 value of 3.86 µM (Gao et al. 2008).
From the stems of Kadsura heteroclita (Schisandraceae, Yunnan, China), dibenzocyclooctadiene lignans were obtained, including kadsulignan K (521), heteroclitins I (523) and J (524), acetoxyl oxokadsurane (525), benzoyl oxokadsurane (526), interiorin B (527), interiorin (451), heteroclitin D (453) and kadsurin (454). Compounds 451 and 527 exhibited anti-HIV-1 activity with EC50 values of 3.3 and 2.9 µM, and therapeutic index values of 52.9 and 65.9, respectively (Pu et al. 2008b).
Twelve new dibenzocyclooctadiene lignans, marlignans A-L (558–573), were obtained from the leaves and stems of Schisandra wilsoniana (Yunnan, China). Compounds 562–564, 567–569 showed anti-HIV-1 activities with EC50 values of 3.6, 3.3, 4.1, 4.1, 4.7, and 3.5 μM and the TI values of 8.7, 15.6, 6.7, 9.3, 7.7, and 16.4, respectively (Yang et al. 2010c). Two new dibenzocyclooctadiene ligans, schinegllignans A and B (613, 614), were isolated from the fruits of Schisandra neglecta (Schisandraceae, Yunnan, China). Compounds 613 showed anti-HIV-1 activities in C8166 cells with a EC50 value of 4.6 and TI value of 18.5 (Duan et al. 2011).
From the stems of Schisandra neglecta (Schisandraceae, Sichuan, China), the new lignans neglignans A-B (590, 591) and E–G (592–594) and the known compounds rubschisantherin (583), gomisins D (585), T (595), F (596), angeloylgomisin Q (597) and schisphenin F (598) were obtained. Copmounds 583, 585, and 590–598 showed anti-HIV-1 activities with EC50 values of 4.7, 9.8, 2.2, 1.4, 5.9, 3.5, 8.2, 8.2, 8.3, 11.5 and 8.3 μM, and TI values of 18.2, 18.9, 40.3, 55.3, > 33.7, > 58.0, 18.1, 15.1, 7.5, 7.4 and 17.8, respectively (Gao et al. 2013).
Norlignans and other lignoids
Norlignans are structures formed by the conjunction of two phenylpropanoid units followed by loss of one carbon from the skeleton. This section covers these compounds, as well as those formed from two phenylpropanoid units, but have undergone skeletal rearrangements or cleavages, termed lignoids. This includes secolignans as well as structures that have seemingly had one of the propyl side-chains completely removed.
During the time covered by this review, 32 norlignans and lignoids isolated from plants were tested for antiviral activity (599–630) (see Table 1 for source plants, antiviral activities, and references). Of the plant-derived norlignans and lignoids, only compounds 605 and 612 displayed antiviral activity at concentrations at or below 1 μM. We found no evidence of synthetic norlignans or lignoids bioassayed for antiviral activity.
Three 7,8-secolignans, marphenols A and B (605–606) together with 7,8-secoholostylone B (607), were isolated from the stems of Schisandra wilsoniana (Schisandraceae, Yunnan, China). Compounds 605–607 inhibited HIV-1IIIB-induced syncytia formation with EC50 values of 1.5, 4.5, 5.4 μM and TI values of 18.27, 4.33 and 9.19, respectively. In a further study of the anti-HIV-1 activity, 605 reduced p24 production in acute HIV-1IIIB-infected C8166 cells with an EC50 value of 9.0 μM, and inhibited primary isolate HIV-1TC-2 replication in PBMCs with an EC50 value of 1.4 μM. However, the compound did not inhibit p24 expression and had no effects on cell to cell fusion in chronically infected H9 cells (Zhang et al. 2010).
(1S)–4–Hydroxy–3–[2–(4–hydroxyl–3–methoxy–phenyl)–1–hydroxymethyl–2–oxo–ethyl]–5–methoxy–benzaldehyde (612) was isolated from the seeds of Herpetospermum caudigerum (Cucurbitaceae, Sichuan, China) and displayed inhibitory activity against HBV on HBsAg and HBeAg secretions with EC50 values of 0.34 and 4.83 × 10–4 μM, respectively, and low cytotoxicity against HepG 2.2.15 cell line with a CC50 value of 2.96 × 105 μM (Yu et al. 2014).
Disscussion
Hundreds of lignans with anti-viral activities were reported from 66 different plant species, belonging to 43 genera, and 34 different families (Table 1). Among them, the genera Kadsura, Phyllanthus and Schisandra contained numerous active lignans with anti-HIV and anti-HBV activities.
Of the 630 lignans and lignoids covered in this review, 153 of them were arylnaphthalene lignans, 73 were aryltetralin lignans, 22 were dibenzylbutyrolactone lignans, 39 were dibenzylbutane lignans, 104 were tetrahydrofuranoid and tetrahydrofurofuranoid lignans, 26 were benzofuran lignans, 24 were neolignans, 157 were dibenzocyclooctadiene lignans and homolignans, and 32 were norlignans and other lignoids. Although aryltetralin and arylnaphthalene lignans have received the most attention in regards to synthesis, and structure–activity-relationship, due in large part to the clinical and commercial success of podophyllotoxin and the promising results of diphyllin (11) and helioxanthin (32), the other classes of lignans and lignoids are also deserving of increased scientific exploration as antiviral lead compounds (Table 2).
Table 2.
Class | #a | # Natural | #(%) active at 1 μM or lessb | #(%) active at 5 μM or lessb | # Syntheticc |
---|---|---|---|---|---|
Arylnaphthalene | 153 | 17 | 5 (29%) | 6 (35%) | 136 |
Aryltetralin | 73 | 31 | 0 (0%) | 3 (10%) | 42 |
Dibenzylbutyrolactone | 22 | 22 | 4 (18%) | 7 (32%) | 0 |
Dibenzylbutane | 39 | 29 | 0 (0%) | 2 (7%) | 10 |
Tetrahydrofuranoid and tetrahydrofurofuranoid | 104 | 104 | 5 (5%) | 9 (9%) | 0 |
Benzofuran | 26 | 26 | 0 (0%) | 0 (0%) | 0 |
Neolignans | 24 | 19 | 0 (0%) | 3 (16%) | 5 |
Dibenzocyclooctadiene lignans and homolignans | 157 | 157 | 1 (1%) | 20 (13%) | 0 |
Norlignans and other lignoids | 32 | 32 | 2 (6%) | 8 (25%) | 0 |
aNumber of compounds in each class of lignan that have been tested for antiviral activity since 1998, and therefore have been included in this review
bThe data was rounded to one significant figure to account for differences in accuracy of reported values. Only natural compounds tested for antiviral activities between 1998 and 2020 are included
cNumber of synthetic compounds using the core structure of each class of lignan that have been tested for antiviral activity since 1998, and therefore have been included in this review
There remain multitudes of lignans and lignoids in all of the categories that have not been tested for antiviral activity, an endeavor that could reveal additional potent potential drugs. Further standardization of antiviral assays could also be beneficial in doing direct comparisons and for SAR studies (a poignant example is compound 627, which has two EC50 values against HIV reported, one 44.68 μM, and one 104 μM).
Between 1963 and 2020, about 200 antiviral drugs were approved for treatment of viral infection. Most of them came from chemical synthesis or structural modifications of natural products (Clercq and Li 2016; Chaudhuri et al. 2018; FDA 2018, 2019, 2020). Although the isolated natural products normally exhibited moderate to weak antiviral activities, some did show excellent antiviral activities. For example, patentiflorin A (153), the 6-deoxyglucoside derivatives of diphyllin, which were isolated from Justicia plants, and shown to potently inhibit a broad spectrum of HIV-1 strains including some resistant strains with EC50 values in the range of 15–37 nM (AZT: 77–95 nM) and mosquito-borne flavivirus such as ZIKV, DENV1, TBEV, WNV, JEV and EBV with EC50 values ranged between 0.12–1.0 μM (Zhang et al. 2017a, 2017b; Martinez-Lopez et al. 2019). Helioxanthin (32) is an arylnaphthalene type lignan obtained from the roots of Heliopsis scabra Dunal (Asteraceae, Taiwan, China) and the aerial parts of Taiwania cryptomerioides Hayata (Taxodiaceae, Taiwan, China). It caught the attention of chemists because of its potent inhibition against HBV gene expression in vitro with an EC50 value of 1 μM and an ID50 value of 31 μM against CEM cell line (Yeo et al. 2005). In a study focusing on the molecular mode of action of 32 on HBV gene expression, this compound was found to suppress the surface antigen promoter (SP) II and the core promoter (CP) selectively, but it had no effect on SPI or the promoter for the X gene. In addition, the suppressive effects on both SPII and CP activity were liver-specific. In another study, compound 32 reduced the DNA-binding activity of the nuclear extract of HepA2 cells to the specific cis element of the HBV promoter for the core antigen, including peroxisome proliferator-activated receptors (PPARs), the PPAR binding site, and the transcription factors α-fetoprotein and specificity protein 1 (Sp1). Moreover, the ectopic expression of PPARγ or HNF4α partially reversed the suppression of HBV RNA by 32. Hence, compound 32 may represent a novel class of anti-HBV agents that can selectively modulate the transcriptional machinery of human liver cells to reduce HBV gene expression and replication (Tseng et al. 2008; Li et al. 2005b). More than 100 derivatives of helioxanthin were synthesised. Among them, the lactam 49 and the cyclic hydrazide derivative 59 exhibited significant inhibitory activity against wild mutant HBV with the EC50 values of 0.08 and 0.03 μM, respectively (Yeo et al. 2005). Compound 49 also inhibited HBV against the drug resistant mutants W10 and DM2 with the EC50 values of 0.004 and 0.0003 μM, respectively (Li et al. 2005b; Cheng et al. 2005a; Tseng et al. 2008). These studies revealed that natural lignans are an important source for the discovery of novel antiviral agents and should be further exploited as lead compounds. Since many lignans have demonstrated antiviral activities, extensive exploration of them through phytochemical studies, chemical synthesis, structure modification, structure–activity relationship analysis and mechanism of action studies could bring next generation of antiviral drugs.
Authors’ contributions
X. X. and D. W. carried out the collection and analysis of data, design of charts and drafted the manuscript. Y. L. participated in revision of manuscript. S. D. and H. Z. came up with the idea of review, edited, and implemented the revision of the manuscripts.
Funding
This work was supported by the Research Grants Council of the Hong Kong Special Administrative Region, China (Nos. HKBU12103618 and HKBU12103917), the Innovation and Technology Commission of Hong Kong Special Administrative Region, China (MHP/105/19), the Research Grants Council of the Hong Kong Baptist University (HKBU), Interdisciplinary Research Matching Scheme (No. RC-IRMS/15-16/02), the Hong Kong Scholars Program Foundation (No. XJ2015047), and Mr. Kwok Yat Wai and Madam Kwok Chung Bo Fun Graduate School Development Fund (WANG Dong-Ying).
Declarations
Conflict of interest
The authors declared that they have no conflicts of interest.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Xin-Ya Xu and Dong-Ying Wang contributed equally.
Contributor Information
Stephen T. Deyrup, Email: sdeyrup@siena.edu
Hong-Jie Zhang, Email: zhanghj@hkbu.edu.hk.
References
- Akram M, Tahir IM, Shah SMA, Mahmood Z, Altaf A, Ahmad K, Munir N, Daniyal M, Nasir S, Mehboob H. Antiviral potential of medicinal plants against HIV, HSV, influenza, hepatitis, and coxsackievirus: a systematic review. Phytother Res. 2018;32:811–822. doi: 10.1002/ptr.6024. [DOI] [PubMed] [Google Scholar]
- Asano J, Chiba K, Tada M, Yoshii T. Antiviral activity of lignans and their glycosides from Justicia procumbens. Phytochem. 1996;42:713–717. doi: 10.1016/0031-9422(96)00024-6. [DOI] [PubMed] [Google Scholar]
- Bar-On Y, Gruell H, Schoofs T, Pai JA, Nogueira L, Butler AL, Millard K, Lehmann C, Suárez I, Oliveira TY, Karagounis T, Cohen YZ, Wyen C, Scholten S, Handl L, Belblidia S, Dizon JP, Vehreschild JJ, Witmer-Pack M, Shimeliovich I, Jain K, Fiddike K, Seaton KE, Yates NL, Horowitz J, Gulick RM, Pfeifer N, Tomaras GD, Seaman MS, Fätkenheuer G, Caskey M, Klein F, Nussenzweig MC. Safety and antiviral activity of combination HIV-1 broadly neutralizing antibodies in viremic individuals. Nat Med. 2018;24:1701–1707. doi: 10.1038/s41591-018-0186-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barquero AA, Dávola ME, Riva DA, Mersich SE, Alché LE. Naturally occurring compounds elicit HIV-1 replication in chronically infected promonocytic cells. BioMed Res Int. 2014;2014:989101. doi: 10.1155/2014/989101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bekhit AED, Bekhit AA. Natural antiviral compounds. Stud Nat Prod Chem. 2014;42:195–228. doi: 10.1016/B978-0-444-63281-4.00007-0. [DOI] [Google Scholar]
- Bertol JW, Santos PR. Antiviral activity of fractions from leaves of Piper regnelli var. pallescens. Rev Bras Farmcogn. 2012;22:1290–1294. [Google Scholar]
- Cao JL, Xue JJ, He SQ, Yang GY, Hu QF. Arylnaphthalene Lignans from Daphne acutiloba Rehd. Asian J Chem. 2010;22:6509–6512. [Google Scholar]
- Castro MA, del Corral JMM, Gordaliza M, Gómez-Zurita MA, de la Puente ML, Betancur-Galvis LA, Sierra J, Feliciano AS. Synthesis, cytotoxicity and antiviral activity of podophyllotoxin analogues modified in the E-ring. Eur J Med Chem. 2003;38:899–911. doi: 10.1016/j.ejmech.2003.05.001. [DOI] [PubMed] [Google Scholar]
- Chang CW, Lin MT, Lee SS, Chen Liu KCS, Hsu FL, Lin JY. Differential inhibition of reverse transcriptase and cellular DNA polymerase-α activities by lignans isolated from Chinese herbs, Phyllanthus myrtifolius Moon, and tannins from Lonicera japonica Thunb and Castanopsis hystrix. Antivir Res. 1995;27:367–374. doi: 10.1016/0166-3542(95)00020-M. [DOI] [PubMed] [Google Scholar]
- Charlton JL. Antiviral activity of lignans. J Nat Prod. 1998;61:1447–1451. doi: 10.1021/np980136z. [DOI] [PubMed] [Google Scholar]
- Chaudhuri S, Symons JA, Deval J. Innovation and trends in the development and approval of antiviral medicines: 1987–2017 and beyond. Antiviral Res. 2018;155:76–88. doi: 10.1016/j.antiviral.2018.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen DF, Zhang SX, Kozuka M, Sun QZ, Feng J, Wang Q, Mukainaka T, Nobukuni Y, Tokuda H, Nishino H, Wang HK, Morris-Natschke SL, Lee KH. Interiotherins C and D, two new lignans from Kadsura interior and antitumor-promoting effects of related neolignans on epstein-barr virus activation. J Nat Prod. 2002;65:1242–1245. doi: 10.1021/np0105127. [DOI] [PubMed] [Google Scholar]
- Chen M, Kilgore N, Lee KH, Chen DF. Rubrisandrins A and B, lignans and related anti-HIV compounds from Schisandra rubriflora. J Nat Prod. 2006;69:1697–1701. doi: 10.1021/np060239e. [DOI] [PubMed] [Google Scholar]
- Chen SW, Wang YH, Jin Y, Tian X, Zheng YT, Luo DQ, Tu YQ. Synthesis and anti-HIV-1 activities of novel podophyllotoxin derivatives. Bioorg Med Chem Lett. 2007;17:2091–2095. doi: 10.1016/j.bmcl.2006.11.070. [DOI] [PubMed] [Google Scholar]
- Chen W, Zhao Y, Bian XW. Inhibitory effect of nordy on HPV16 E6 gene in human immortalized endocervical cells. Chin J Cancer Res. 2008;20:1–4. doi: 10.1007/s11670-008-0001-8. [DOI] [Google Scholar]
- Chen QJ, Ouyang MA, Tan QW, Zhang ZK, Wu ZJ, Lin QY. Constituents from the seeds of Brucea javanica with inhibitory activity of Tobacco mosaic virus. J Asian Nat Prod Res. 2009;11:539–547. doi: 10.1080/10286020902932708. [DOI] [PubMed] [Google Scholar]
- Chen H, Li J, Wu Q, Niu XT, Tang MT, Guan XL, Li J, Yang RY, Deng SP, Su XJ. Anti-HBV activities of Streblus asper and constituents of its roots. Fitoterapia. 2012;83:643–649. doi: 10.1016/j.fitote.2012.01.009. [DOI] [PubMed] [Google Scholar]
- Chen HW, Cheng JX, Liu MT, King K, Peng JY, Zhang XQ, Wang CH, Shresta S, Schooley RT, Liu YT. Inhibitory and combinatorial effect of diphyllin, a v-ATPase blocker, on influenza viruses. Antivir Res. 2013;99:371–382. doi: 10.1016/j.antiviral.2013.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng YC, Ying CX, Leung CH, Li Y. New targets and inhibitors of HBV replication to combat drug resistance. J Clin Virol. 2005;34:S147–S150. doi: 10.1016/S1386-6532(05)80026-5. [DOI] [PubMed] [Google Scholar]
- Cheng MJ, Lee KH, Tsai IL, Chen IS. Two new sesquiterpenoids and anti-HIV principles from the root bark of Zanthoxylum ailanthoides. Bioorgan Med Chem. 2005;13:5915–5920. doi: 10.1016/j.bmc.2005.07.050. [DOI] [PubMed] [Google Scholar]
- Clercq ED, Li G. Approved antiviral drugs over the past 50 years. Clin Microbiol Rev. 2016;29:695–747. doi: 10.1128/CMR.00102-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cragg GM, Newman DJ. Plants as source of anti-cancer agents. J Ethnopharmacol. 2005;100:72–79. doi: 10.1016/j.jep.2005.05.011. [DOI] [PubMed] [Google Scholar]
- Cragg GM, Kingston DGI, Newman DJ. Anticancer agents from natural products. 2. Boca Raton: CRC Press; 2011. [Google Scholar]
- Craigo J, Callahan M, Huang RCC, DeLucia AL. Inhibition of human papillomavirus type 16 gene expression by nordihydroguaiaretic acid plant lignan derivatives. Antivir Res. 2000;47:19–28. doi: 10.1016/S0166-3542(00)00089-9. [DOI] [PubMed] [Google Scholar]
- Cui H, Xu B, Wu TZ, Xu J, Yuan Y, Gu Q. Potential antiviral lignans from the roots of Saururus chinensis with activity against epstein-barr virus lytic replication. J Nat Prod. 2014;77:100–110. doi: 10.1021/np400757k. [DOI] [PubMed] [Google Scholar]
- Cui QH, Du RK, Liu MM, Rong LJ. Lignans and their derivatives from plants as antivirals. Molecules. 2020;25:183. doi: 10.3390/molecules25010183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duan YX, Cao JL, Wen RR, Yang GY, Pu JX, Sun HD, Xiao WL, Li GP. Dibenzocyclooctadiene lignans from Schisandra neglecta and their anti-HIV-1 activities. J Asian Nat Prod Res. 2011;13:592–598. doi: 10.1080/10286020.2011.576843. [DOI] [PubMed] [Google Scholar]
- Fan P, Zhang X, Li T, Dai Y, Li GP, Hu QF. A new lignan from the fruit of Schisandra propinqua var. propinqua and its anti-HIV activity. Asian J Chem. 2010;22:1075–1078. [Google Scholar]
- FDA (U.S. Food and Drug Administration) (2018). Novel drug approvals for 2018. https://www.fda.gov/drugs/new-drugs-fda-cders-new-molecular-entities-and-new-therapeutic-biological-products/novel-drug-approvals-2018; accessed on March 21, 2021
- FDA (U.S. Food and Drug Administration) (2019). Novel drug approvals for 2019. https://www.fda.gov/drugs/new-drugs-fda-cders-new-molecular-entities-and-new-therapeutic-biological-products/novel-drug-approvals-2019; accessed on March 21, 2021
- FDA (U.S. Food and Drug Administration) (2020). Novel drug approvals for 2020. https://www.fda.gov/drugs/new-drugs-fda-cders-new-molecular-entities-and-new-therapeutic-biological-products/novel-drug-approvals-2020; accessed on March 21, 2021
- Gao XM, Pu JX, Huang SX, Yang LM, Huang H, Xiao WL, Zheng YT, Sun HD. Lignans from Kadsura angustifolia. J Nat Prod. 2008;71:558–563. doi: 10.1021/np0705108. [DOI] [PubMed] [Google Scholar]
- Gao XM, Li XS, Yang XZ, Mu HX, Chen YK, Yang GY, Hu QF. Lignan derivatives from the leaves Nicotiana tabacum and their activities. Heterocycles. 2012;85:147–153. doi: 10.3987/COM-11-12349. [DOI] [Google Scholar]
- Gao XM, Wang RR, Niu DY, Meng CY, Yang LM, Zheng YT, Yang GY, Ju QF, Sun HD, Xiao WL. Bioactive dibenzocyclooctadiene lignans from the stems of Schisandra neglecta. J Nat Prod. 2013;76:1052–1057. doi: 10.1021/np400070x. [DOI] [PubMed] [Google Scholar]
- Guan Y, Wang D, Tan GT, Hung NV, Cuong NM, Pezzuto JM, Fong HHS, Soejarto DD, Zhang H. Litsea species as potential antiviral plant sources. Am J Chin Med. 2016;44:275–290. doi: 10.1142/S0192415X16500166. [DOI] [PubMed] [Google Scholar]
- Halim TA, Farooqi AA, Zaman F. Nip the HPV encoded evil in the cancer bud: HPV reshapes TRAILs and signaling landscapes. Cancer Cell Int. 2013;13:1–18. doi: 10.1186/1475-2867-13-61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han YS, Xiao WL, Xu HT, Kramer VG, Quan YD, Mesplède T, Oliveira M, Colby-Germinario SP, Sun HD, Wainberg MA. Identification of a dibenzocyclooctadiene lignan as a HIV-1 non-nucleoside reverse transcriptase inhibitor. Antivir Chem Chemoth. 2015;24:28–38. doi: 10.1177/2040206614566580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hande KR. Etoposide: four decades of development of a topoisomerase II inhibitor. Eur J Cancer. 1998;34:1514–1521. doi: 10.1016/S0959-8049(98)00228-7. [DOI] [PubMed] [Google Scholar]
- Hayashi K, Narutaki K, Nagaoka Y, Hayashi T, Uesato S. Therapeutic effect of arctiin and arctigenin in immunocompetent and immunocompromised mice infected with influenza A virus. Biol Pharm Bull. 2010;33:1199–1205. doi: 10.1248/bpb.33.1199. [DOI] [PubMed] [Google Scholar]
- Hoang VD, Tan GT, Zhang HJ, Tamez PA, Hung NV, Cuong NM, Soejarto DD, Fong HHS, Pezzuto JM. Natural anti-HIV agents-part I: (+)-demethoxyepiexcelsin and vertivillatol from Litsea vertivillata. Phytochemistry. 2002;59:325–329. doi: 10.1016/S0031-9422(01)00454-X. [DOI] [PubMed] [Google Scholar]
- Hu QF, Mu HX, Huang HT, Lv HY, Li SL, Tu PF, Li GP. Secolignans, neolignans and phenylpropanoids from Daphne feddei and their biological activities. Chem Pharm Bull. 2011;59:1421–1424. doi: 10.1248/cpb.59.1421. [DOI] [PubMed] [Google Scholar]
- Huang RL, Huang YL, Ou JC, Chen CC, Gsu FL, Chang CM. Screening of 25 compounds isolated from Phyllanthus Species for anti-human hepatitis B virus in vitro. Phytother Res. 2003;17:449–453. doi: 10.1002/ptr.1167. [DOI] [PubMed] [Google Scholar]
- Huang RCC, Li Y, Giza PE, Gnabre JN, Abd-Elazem IS, King KY, Hwu JR. Novel antiviral agent tetraglycylated nordihydroguaiaretic acid hydrochloride as a host-dependent viral inhibitor. Antivir Res. 2003;58:57–64. doi: 10.1016/S0166-3542(02)00189-4. [DOI] [PubMed] [Google Scholar]
- Huang SZ, Zhang XJ, Li XY, Jiang HZ, Ma QY, Wang PC, Liu YQ, Hu JM, Zheng YT, Zhou J, Zhao YX. Phenols with anti-HIV activity from Daphne acutiloba. Planta Med. 2012;78:182–185. doi: 10.1055/s-0031-1280263. [DOI] [PubMed] [Google Scholar]
- Hussain M, Galvin HD, Haw TY, Nutsford AN, Hussain M. Drugs resistance in influenza A virus: the epidemiology and management. Infect Drug Resist. 2017;10:121–134. doi: 10.2147/IDR.S105473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hwu JR, Hsu MH, Huang RCC. New nordihydroguaiaretic acid derivatives as anti-HIV agents. Bioorg Med Chem Lett. 2008;18:1884–1888. doi: 10.1016/j.bmcl.2008.02.018. [DOI] [PubMed] [Google Scholar]
- Ishida J, Wang HK, Oyama M, Cosentino ML, Hu CQ, Lee KH. Anti-AIDS agents. 46. Anti-HIV activity of harman, an anti-HIV principle from Symplocos setchuensis, and its derivatives. J Nat Prod. 2001;64:958–960. doi: 10.1021/np0101189. [DOI] [PubMed] [Google Scholar]
- Ito C, Itoiqawa M, Oqata M, Mou XY, Tokuda H, Nishino H, Furukaw H. Lignans as anti-tumor-promoter from the seeds of Hernandia ovigera. Planta Med. 2001;67:166–168. doi: 10.1055/s-2001-11501. [DOI] [PubMed] [Google Scholar]
- Izzedine H, Launay-Vacher V, Deray G. Antiviral drug-induced nephrotoxicity. Am J Kidney Dis. 2005;45:804–817. doi: 10.1053/j.ajkd.2005.02.010. [DOI] [PubMed] [Google Scholar]
- Janmanchi D, Tseng YP, Wang KC, Huang RL, Lin CH, Yeh SF. Synthesis and the biological evaluation of arylnaphthalene lignans as anti-hepatitis B virus agents. Bioorgan Med Chem. 2010;18:1213–1226. doi: 10.1016/j.bmc.2009.12.038. [DOI] [PubMed] [Google Scholar]
- Janmanchi D, Lin CH, Hsieh JY, Tseng YP, Chen TA, Jhuang HJ, Yeh SF. Synthesis and biological evaluation of helioxanthin analogues. Bioorgan Med Chem. 2013;21:2163–2176. doi: 10.1016/j.bmc.2012.11.037. [DOI] [PubMed] [Google Scholar]
- Kaplan IW. Condylomata acuminate. New Orleans Med Surg J. 1942;94:388–390. [Google Scholar]
- Kim HJ, Yu YG, Park H, Lee YS. HIV gp41 binding phenolic compounents from Fraxinus sieboldiana var. angustata. Planta Med. 2002;68:1034–1036. doi: 10.1055/s-2002-35665. [DOI] [PubMed] [Google Scholar]
- Konigheim BS, Goleniowski ME, Contigiani MS. Cytotoxicity and antiviral activity of a lignan extracted from Larrea divaricate. Drug Des Rev. 2005;2:81–83. [Google Scholar]
- Kuo YH, Li SY, Wu MD, Huang RL, Yang Kuo LM, Chen CF. A New anti-HBeAg lignan, kadsumarin A, from Kadsura matsudai and Schizandra arisanensis. Chem Pharm Bull. 1999;47:1047–1048. doi: 10.1248/cpb.47.1047. [DOI] [PubMed] [Google Scholar]
- Kuo YH, Li SY, Huang RL, Wu MD, Huang HC, Lee KH. Schizarin B, C, D, and E, four new lignans from Kadsura matsudai and their antihepatitis activities. J Nat Prod. 2001;64:487–490. doi: 10.1021/np000261m. [DOI] [PubMed] [Google Scholar]
- Kuo YH, Wu MD, Huang RL, Yang Kuo LM, Hsu YW, Liaw CC, Hung CC, Shen YC, Ong CW. Antihepatitis activity (anti-HBsAg and anti-HBeAg) of C19 homolignans and six novel C18 dibenzocyclooctadiene lignans from Kadsura japonica. Planta Med. 2005;71:646–653. doi: 10.1055/s-2005-871271. [DOI] [PubMed] [Google Scholar]
- Kuo YC, Kuo YH, Lin YL, Tsai WJ. Yatein from Chamaecyparis obtusa suppresses herpes simplex virus type 1 replication in HeLa cells by interruption the immediate-early gene expression. Antivir Res. 2006;70:112–120. doi: 10.1016/j.antiviral.2006.01.011. [DOI] [PubMed] [Google Scholar]
- Lan KH, Wang YW, Lee WP, Lan KL, Tseng SH, Hung LH, Yen SH, Lin HC, Lee SD. Multiple effects of honokiol on the life cycle of hepatitis C virus. Liver Int. 2012;32:989–997. doi: 10.1111/j.1478-3231.2011.02621.x. [DOI] [PubMed] [Google Scholar]
- Lee DG, Jin Q, Jin HG, Shin JE, Choi EJ, Woo ER. Isolation of virus-cell fusion inhibitory components from the stem bark of Styrax japonica S. et Z Arch. Pharm Res. 2010;33:863–866. doi: 10.1007/s12272-010-0608-4. [DOI] [PubMed] [Google Scholar]
- Lee J, Huh MS, Kim YC, Hattori M, Otake T. Lignan, sesquilignans and dilignans, novel HIV-1 protease and cytopathic effect inhibitors purified from the rhizomes of Saururus chinensis. Antivir Res. 2010;85:425–428. doi: 10.1016/j.antiviral.2009.11.002. [DOI] [PubMed] [Google Scholar]
- Lei C, Huang SX, Chen JJ, Pu JX, Yang LB, Zhao Y, Liu JP, Gao XM, Xiao WL, Sun HD. Lignans from Schisandra propinqua var. propinqua. Chem Pharm Bull. 2007;55:1281–1283. doi: 10.1248/cpb.55.1281. [DOI] [PubMed] [Google Scholar]
- Li C, Dai Y, Zhang SX, Duan YH, Liu ML, Chen LY, Yao XS. Quinoid glycosides from Forsythia suspensa. Phytochemistry. 2014;104:105–113. doi: 10.1016/j.phytochem.2014.04.010. [DOI] [PubMed] [Google Scholar]
- Li SY, Wu MD, Wang CW, Kuo YH, Huang RL, Lee KH. A novel anti-HBeAg homolignan, taiwanschirin D from Kadsura matsudai. Chem Pharm Bull. 2000;48:1992–1993. doi: 10.1248/cpb.48.1992. [DOI] [PubMed] [Google Scholar]
- Li RT, Han QB, Zheng YT, Wang RR, Yang LM, Peng LY, Xiao WL, Sun HD. Anti-HIV lignans from Schisandra micrantha. Chin J Nat Med. 2005;3:208–212. [Google Scholar]
- Li YS, Wang ZT, Zhang M, Luo SD, Chen JJ. A new pinoresinol-type lignan from Ligularia kanaitizensis. Nat Prod Res. 2005;19:125–129. doi: 10.1080/14786410410001704723. [DOI] [PubMed] [Google Scholar]
- Li Y, Fu L, Yeo H, Zhu JL, Chou CK, Kou SF, Gullen E, Austin D, Cheng YC. Inhibition of hepatitis B virus gene expression and replication by helioxanthin and it derivative Antivir. Chem Chemoth. 2005;16:193–201. doi: 10.1177/095632020501600305. [DOI] [PubMed] [Google Scholar]
- Li HM, Luo YM, Pu JX, Li XN, Lei C, Wang RR, Zheng YT, Sun HD, Li RT. Four new dibenzocyclooctadiene lignans from Schisandra rubriflora. Helv Chim Acta. 2008;91:1053–1062. doi: 10.1002/hlca.200890113. [DOI] [Google Scholar]
- Li YK, Ma YH, Peng YF, Dai Y, Li GP. A new lignan from the fruit of Schisandra lancifolia and its anti-HIV activity. Asian J Chem. 2009;21:5794–5796. [Google Scholar]
- Li XN, Pu JX, Du X, Yang LM, An HM, Lei C, He F, Luo X, Zheng YT, Lu Y, Xiao WL, Sun HD. Lignans with anti-HIV activity from Schisandra propinqua var. sinensis. J Nat Prod. 2009;72:1133–1141. doi: 10.1021/np900123z. [DOI] [PubMed] [Google Scholar]
- Li YR, Cheng W, Zhu CG, Yao CS, Xiong L, Tian Y, Wang SJ, Lin S, Hu JF, Yang YC, Guo Y, Yang Y, Li Y, Yuan YH, Chen NH, Shi JG. Bioactive neolignans and lignans from the bark of Machilus robusta. J Nat Prod. 2011;74:1444–1452. doi: 10.1021/np2001896. [DOI] [PubMed] [Google Scholar]
- Li J, Huang Y, Guan XL, Li J, Deng SP, Wu Q, Zhang YJ, Su XJ, Yang RY. Anti-hepatitis B virus constituents from the stem bark of Streblus asper. Phytochemistry. 2012;82:100–109. doi: 10.1016/j.phytochem.2012.06.023. [DOI] [PubMed] [Google Scholar]
- Li XN, Lei C, Yang LM, Li HM, Huang SX, Du X, Pu JX, Xiao WL, zheng YT, Sun HD, Three new arylnaphthalene lignans from Schisandra propinqua var. sinensis. Fitoterapia. 2012;83:249–252. doi: 10.1016/j.fitote.2011.11.006. [DOI] [PubMed] [Google Scholar]
- Li LQ, Li J, Huang Y, Wu Q, Deng SP, Su XJ, Yang RY, Huang JG, Chen ZZ, Shan Li. Lignans from the heartwood of Streblus asper and their inhibiting activities to Hepatitis B virus. Fitoterapia. 2012;83:303–309. doi: 10.1016/j.fitote.2011.11.008. [DOI] [PubMed] [Google Scholar]
- Li J, Meng AP, Guan XL, Li J, Wu Q, Deng SP, Su XJ, Yang RY. Anti-hepatitis B virus lignans from the root of Streblus asper. Bioorg Med Chem Lett. 2013;23:2238–2244. doi: 10.1016/j.bmcl.2013.01.046. [DOI] [PubMed] [Google Scholar]
- Li J, Zhou BX, Li CF, Chen QY, Wang YT, Li ZT, Chen TT, Yang CG, Jiang ZH, Zhong NS, Yang ZF, Chen RC. Lariciresinol-4-O-β-d-glucopyranoside from the root of Isatis indigotica inhibits influenza A virus-induced pro-inflammatory response. J Ethnopharmacol. 2015;174:379–386. doi: 10.1016/j.jep.2015.08.037. [DOI] [PubMed] [Google Scholar]
- Liang CQ, Hu J, Luo RH, Shi YM, Shang SZ, Gao ZH, Wang RR, Zheng YT, Xiong WY, Zhang HB, Xiao WL, Sun HD. Six new lignans from the leaves and stems of Schisandra sphenanthera. Fitoterapia. 2013;84:171–177. doi: 10.1016/j.fitote.2013.03.008. [DOI] [PubMed] [Google Scholar]
- Liao Z, Li XS, Lu YL, Shi HL, LiZ HuQF. Norlignans from the roots and stems of nicotiana tabacum and their antitobacco mosaic virus activity. Asian J Chem. 2012;24:4895–4897. [Google Scholar]
- Lin HW, Sun MX, Wang YH, Yang LM, Yang YR, Huang N, Xuan LJ, Xu YM, Bai DL, Zheng YT. Anti-HIV activities of the compounds isolated from Polygonum cuspidatum and Polygonum multiflorum. Planta Med. 2010;76:889–892. doi: 10.1055/s-0029-1240796. [DOI] [PubMed] [Google Scholar]
- Liu ZL, Liu YQ, Zhao L, Xu J, Tian X. The phenylpropanoids of Aster flaccidus. Fitoterapia. 2010;81:140–144. doi: 10.1016/j.fitote.2009.08.004. [DOI] [PubMed] [Google Scholar]
- Liu JF, Jiang ZY, Geng CA, Zhang Q, Shi Y, Ma YB, Zhang XM, Chen JJ. Two new lignans and anti-HBV constituents from Illicium henryi. Chem Biodivers. 2011;8:692–698. doi: 10.1002/cbdv.201000110. [DOI] [PubMed] [Google Scholar]
- Liu S, Wei WX, Shi KC, Cao X, Zhou M, Liu ZP. In vitro and in vivo anti-hepatitis B virus activities of the lignan niranthin isolated from Phyllanthus niruri L. J Ethnopharmacol. 2014;155:1061–1067. doi: 10.1016/j.jep.2014.05.064. [DOI] [PubMed] [Google Scholar]
- Ma WH, Ma XL, Huang H, Zhou P, Chen DF. Dibenzocyclooctane Lignans from the stems of Kadsura induta and their antiviral effect on hepatitis B virus. Chem Biodivers. 2007;4:972–996. doi: 10.1002/cbdv.200790087. [DOI] [PubMed] [Google Scholar]
- Ma WH, Lu Y, Huang H, Zhou P, Chen DF. Schisanwilsonins A-G and related anti-HBV lignans from the fruits of Schisandra wilsoniana. Bioorg Med Chem Lett. 2009;19:4958–4962. doi: 10.1016/j.bmcl.2009.07.078. [DOI] [PubMed] [Google Scholar]
- Ma WH, Lu Y, Chen DF. Dibenzocyclooctane Lignans from the Stems of Schisandra wilsoniana. Planta Med. 2013;79:1051–1055. doi: 10.1055/s-0032-1328747. [DOI] [PubMed] [Google Scholar]
- Martinez JP, Sasse F, Brönstrup M, Diez J, Meyerhans A. Antiviral drug discovery: broad-spectrum drugs from nature. Nat Prod Rep. 2014;32:29–48. doi: 10.1039/C4NP00085D. [DOI] [PubMed] [Google Scholar]
- Martinez-Lopez A, Persaud M, Chavez MP, Zhang HJ, Rong LJ, Liu SF, Wang TT, Sarafianos SG, Diaz-Griffero F. Glycosylated diphyllin as a broad-spectrum antiviral agent against Zika virus. EBioMedicine. 2019;47:269–283. doi: 10.1016/j.ebiom.2019.08.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ming DS, López A, Hillhouse BJ, French CJ, Hudson JB, Towers GHN. Bioactive constituents from Iryanthera megistophylla. J Nat Prod. 2002;65:1412–1416. doi: 10.1021/np020169l. [DOI] [PubMed] [Google Scholar]
- Mu HX, Li XS, Fan P, Yang GY, Pu JX, Sun HD, Hu QF, Xiao WL. Dibenzocyclooctadiene lignans from the fruits of Schisandra rubriflora and their anti-HIV-1 activities. J Asian Nat Prod Res. 2011;13:393–399. doi: 10.1080/10286020.2011.565748. [DOI] [PubMed] [Google Scholar]
- Mukhtar M, Arshad M, Ahmad M, Pomerantz RJ, Wigdahl B, Parveen Z. Antiviral potentials of medicinal plants. Virus Res. 2008;131:111–120. doi: 10.1016/j.virusres.2007.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Newman DJ, Cragg GM. Natural products as sources of new drugs over the nearly four decades from 01/1981 to 09/2019. J Nat Prod. 2020;83:770–803. doi: 10.1021/acs.jnatprod.9b01285. [DOI] [PubMed] [Google Scholar]
- Ouyang MA, Wein YS, Zhang ZK, Kuo YH. Inhibitory activity against tobacco mosaic virus (TMV) replication of pinoresinol and syringaresinol lignans and their glycosides from the root of Rhus javanica var. roxburghiana. J Agric Food Chem. 2007;55:6460–6465. doi: 10.1021/jf0709808. [DOI] [PubMed] [Google Scholar]
- Ovenden SPB, Yu J, Wan SS, Sberna G, Tait RM, Rhodes D, Cox S, Coates J, Walsh NG, Meurer-Grimes BM. Globoidnan A: a lignan from Eucalyptus globoidea inhibits HIV integrase. Phytochemistry. 2004;65:3255–3259. doi: 10.1016/j.phytochem.2004.10.006. [DOI] [PubMed] [Google Scholar]
- Pan WH, Liu KL, Guan YF, Tan GT, Hung NV, Cuong NM, Soejarto DD, Pezzuto JM, Fong HHS, Zhang HJ. Bioactive compounds from Vitex leptobotrys. J Nat Prod. 2014;77:663–667. doi: 10.1021/np400779v. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Panthong K, Srisud Y, Rukachaisirikul V, Hutadilok-Towatana N, Voravuthikunchai SP, Tewtrakul S. Benzene, coumarin and quinolinone derivatives from roots of Citrus hystrix. Phytochemistry. 2013;88:79–84. doi: 10.1016/j.phytochem.2012.12.013. [DOI] [PubMed] [Google Scholar]
- Parhira S, Yang ZF, Zhu GY, Chen QL, Zhou BX, Wang YT, Liu L, Bai LP, Jiang ZH. In vitro anti-influenza virus activities of a new lignan glycoside from the latex of Calotropis gigantean. PLoS ONE. 2014;9:1–13. doi: 10.1371/journal.pone.0104544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park S, Song J-H, Nhiem NX, Ko H-J, Kim SH. The chemical constituents from twigs of Lindera glauca (Siebold & Zucc.) Blume and their antiviral activities. Phytochem Lett. 2018;25:74–80. doi: 10.1016/j.phytol.2018.04.008. [DOI] [Google Scholar]
- Piccinelli AL, Mahmood N, Mora G, Poveda L, De Simone F, Rastrelli L. Anti-HIV activity of dibenzylbutyrolactone-type lignans from Phenax species endemic in Costa Rica. J Pharm Pharmacol. 2005;57:1109–1115. doi: 10.1211/jpp.57.9.0006. [DOI] [PubMed] [Google Scholar]
- Pu JX, Yang LM, Xiao WL, Li RT, Lei C, Gao XM, Huang SX, Li SH, Zheng YT, Huang H. Compounds from Kadsura heteroclita and related anti-HIV activity. Phytochemistry. 2008;69:1266–1272. doi: 10.1016/j.phytochem.2007.11.019. [DOI] [PubMed] [Google Scholar]
- Pu JX, Gao XM, Lei C, Xiao WL, Wang RR, Yang LB, Zhao Y, Li LM, Huang SX, Zheng YT, Sun HD. Three new compounds from Kadsura longipedunculata. Chem Pharm Bull. 2008;56:1143–1146. doi: 10.1248/cpb.56.1143. [DOI] [PubMed] [Google Scholar]
- Pushpa R, Nishant R, Navin K, Pankaj G. Antiviral potential of medicinal plants: an overview. Int Res J Pharm. 2013;4:8–16. doi: 10.7897/2230-8407.04603. [DOI] [Google Scholar]
- Qian XJ, Jin YS, Chen HS, Xu QQ, Ren H, Zhu SY, Tang HL, Wang Y, Zhao P, Qi ZT, Zhu YZ. Trachelogenin, a novel inhibitor of hepatitis C virus entry through CD81. J Gen Virol. 2016;97:1134–1144. doi: 10.1099/jgv.0.000432. [DOI] [PubMed] [Google Scholar]
- Quasdorff M, Protzer U. Control of hepatitis B virus at the level of transcription. J Viral Hepat. 2010;17:527–536. doi: 10.1111/j.1365-2893.2010.01315.x. [DOI] [PubMed] [Google Scholar]
- Rastrelli L, Simone FD, Mora G, Poveda JL, Aquino R. Phenolic constituents of Phenax angustifolius. J Nat Prod. 2001;64:79–81. doi: 10.1021/np000290x. [DOI] [PubMed] [Google Scholar]
- Rumschlag-Booms E, Zhang HJ, Soejarto DD, Fong HHS, Rong L. Development of antiviral screening protocol: one-stone-two-birds. J Antivir Antiretrovir. 2011;3:8–10. doi: 10.4172/jaa.1000027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sagar KS, Chang CC, Wang WK, Lin JY, Lee SS. Preparation and anti-HIV activities of retrojusticidin B analogs and azalignans. Bioorgan Med Chem. 2004;12:4045–4054. doi: 10.1016/j.bmc.2004.05.036. [DOI] [PubMed] [Google Scholar]
- Sawasdee K, Chaowasku T, Lipipun V, Dufat TH, Michel S, Likhitwitayawuid K. New neolignans and a lignan from Miliusa fragrans, and their anti-herpetic and cytotoxic activities. Tetrahedron Lett. 2013;54:4259–4263. doi: 10.1016/j.tetlet.2013.05.144. [DOI] [Google Scholar]
- Shang SZ, Han YS, Shi YM, Du X, Liang CQ, Wainberg MA, Gao ZH, Xiao WL, Sun HD. Four new lignans from the leaves and stems of Schisandra propinqua var. sinensis. Nat Prod Bioprospect. 2013;3:56–60. doi: 10.1007/s13659-013-0017-8. [DOI] [Google Scholar]
- Shang SZ, Chen H, Liang CQ, Gao ZH, Du X, Wang RR, Shi YM, Zheng YT, Xiao WL, Sun HD. Phenolic constituents from Parakmeria yunnanensis and their anti-HIV-1 activity. Arch Pharm Res. 2013;36:1223–1230. doi: 10.1007/s12272-013-0070-1. [DOI] [PubMed] [Google Scholar]
- Shen WD, Zou XP, Chen M, Liu PF, Shen YH, Huang SL, Guo HM, Zhang LL. Effects of diphyllin as a novel V-ATPase inhibitor on gastric adenocarcinoma. Eur J Pharmacol. 2011;667:330–338. doi: 10.1016/j.ejphar.2011.05.042. [DOI] [PubMed] [Google Scholar]
- Shi YM, Zhong WM, Chen H, Wang RR, Shang SZ, Liang CQ, Gao ZH, Zheng YT, Xiao WL, Sun HD. New lignans from the leaves and stems of Schisandra chinensis and their anti-HIV-1 activities. Chin J Chem. 2014;32:734–740. doi: 10.1002/cjoc.201400001. [DOI] [Google Scholar]
- Short KR, Kedzierska K, van de Sandt CE. Back to the future: lessons learned from the 1918 influenza pandemic Front Cell. Infect Microbiol. 2018;8:343. doi: 10.3389/fcimb.2018.00343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Song QY, Zhang CJ, Li Y, Wen J, Zhao XW, Liu ZL, Gao K. Lignans from the fruit of Schisandra sphenanthera, and their inhibition of HSV-2 and adenovirus. Phytochem Lett. 2013;6:174–178. doi: 10.1016/j.phytol.2012.12.008. [DOI] [Google Scholar]
- Soto-Acosta R, Bautista-Carbajal P, Syed GH, Siddiqui A, Del Angel RM. Nordihydroguaiaretic acid (NDGA) inhibits replication and viral morphogenesis of dengue virus. Antivir Res. 2014;109:132–140. doi: 10.1016/j.antiviral.2014.07.002. [DOI] [PubMed] [Google Scholar]
- Sun QZ, Chen DF, Ding PL, Ma CM, Kakuda H, Nakamura N, Hattori M. Three new lignans, longipedunins A-C, from Kadsura longipedunculata, and their inhibitory activity against HIV-1 protease. Chem Pharm Bull. 2006;54:129–132. doi: 10.1248/cpb.54.129. [DOI] [PubMed] [Google Scholar]
- Sun X, Ma G, Zhang D, Huang W, Ding G, Hu H, Tu G, Guo B. New lignans and iridoid glycosides Fromdipsacus asper Wall. Molecules. 2015;20:2165–2175. doi: 10.3390/molecules20022165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Syed GH, Siddiqui A. Effects of hypolipidemic agent nordihydroguaiaretic acid on lipid droplets and Hepatitis C virus. Hepatology. 2011;54:1936–1946. doi: 10.1002/hep.24619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tan QW, Ou Yang MA, Wu ZJ. A new seco-neolignan glycoside from the root bark of Ailanthus altissima. Nat Prod Res. 2012;26:1375–1380. doi: 10.1080/14786419.2011.587187. [DOI] [PubMed] [Google Scholar]
- Tanaka T, Ikeda T, Kaku M, Zhu XH, Okawa M, Yokomizo K, Uyeda M, Nohara T. A New Lignan Glycoside and Phenylethanoid Glycosides from Strobilanthes cusia Bremek. Chem Pharm Bull. 2004;52:1242–1245. doi: 10.1248/cpb.52.1242. [DOI] [PubMed] [Google Scholar]
- Teponno RB, Kusari S, Spiteller M. Recent advances in research on lignans and neolignans. Nat Prod Rep. 2016;33:1044–1092. doi: 10.1039/C6NP00021E. [DOI] [PubMed] [Google Scholar]
- Tseng YP, Kuo YH, Hu CP, Jeng KS, Janmanchi D, Lin CH, Chou CK, Yeh SF. The role of helioxanthin in inhibiting human hepatitis B viral replication and gene expression by interfering with the host transcriptional machinery of viral promoters. Antivir Res. 2008;77:206–214. doi: 10.1016/j.antiviral.2007.12.011. [DOI] [PubMed] [Google Scholar]
- Tuchinda P, Kornsakulkarn J, Pohmakotr M, Kongsaeree P, Prabpai S, Yoosook C, Kasisit J, Napaswad C, Sophasan S, Reutrakul V. Dichapetalin-type triterpenoids and lignans from the aerial parts of Phyllanthus acutissima. J Nat Prod. 2008;71:655–663. doi: 10.1021/np7007347. [DOI] [PubMed] [Google Scholar]
- Wang GK, Lin BB, Rao R, Zhu K, Qin XY, Xie GY, Qin MJ. A new lignan with anti-HBV activity from the roots of Bombax ceiba. Nat Prod Res. 2013;27:1348–1352. doi: 10.1080/14786419.2012.740032. [DOI] [PubMed] [Google Scholar]
- Wangteeraprasert R, Lipipun V, Gunaratnam M, Neidle S, Gibbons S, Likhitwitayawuid K. Bioactive compounds from Carissa spinarum. Phytother Res. 2012;26:1496–1499. doi: 10.1002/ptr.4607. [DOI] [PubMed] [Google Scholar]
- Wei WX, Li XR, Wang KW, Zheng ZW, Zhou M. Lignans with anti-hepatitis B virus activities from Phyllanthus niruri L. Phytother Res. 2012;26:964–968. doi: 10.1002/ptr.3663. [DOI] [PubMed] [Google Scholar]
- Wen CC, Kuo YH, Jan JT, Liang PH, Wang SY, Liu HG, Lee CK, Chang SZ, Kuo CJ, Lee SS, Hou CC, Hsiao PW, Chien SC, Shyur LC, Yang NS. Specific plant terpenoids and lignoids possess potent antiviral activities against severe acute respiratory syndrome coronavirus. J Med Chem. 2007;50:4087–4095. doi: 10.1021/jm070295s. [DOI] [PubMed] [Google Scholar]
- WHO (World Health Organization). HIV data and statistics. https://www.who.int/news-room/fact-sheets/detail/hiv-aids/. Accessed on 21 Mar 2021
- Worldometer. COVID-19 coronavirus pandemic. https://www.worldometers.info/coronavirus/; accessed on Mar 21, 2021
- Wu Z, McGoogan JM. Characteristics of and important lessons from the coronavirus disease 2019 (COVID-19) outbreak in China: summary of a report of 72 314 cases from the Chinese Center for Disease Control and Prevention. JAMA. 2020;323:1239–1242. doi: 10.1001/jama.2020.2648. [DOI] [PubMed] [Google Scholar]
- Wu MD, Huang RL, Yang Kuo LM, Hung CC, Ong CW, Kuo YH. The anti-HBsAg (human type B hepatitis, surface antigen) and Anti-HBeAg (human type B hepatitis, e antigen) C18 dibenzocyclooctadiene lignans from Kadsura matsudai and Schizandra arisanensis. Chem Pharm Bull. 2003;51:1233–1236. doi: 10.1248/cpb.51.1233. [DOI] [PubMed] [Google Scholar]
- Wu SF, Lin CK, Chuang YS, Chang FR, Tseng CK, Wu YC, Lee JC. Anti-hepatitis C virus activity of 3-hydroxy caruilignan C from Swietenia macrophylla stems. J Viral Hepatitis. 2012;19:364–370. doi: 10.1111/j.1365-2893.2011.01558.x. [DOI] [PubMed] [Google Scholar]
- Xiao WL, Huang SX, Wang RR, Zhong JL, Gao XM, He F, Pu JX, Lu Y, Zheng YT, Zheng QT, Sun HD. Nortriterpenoids and lignans from Schisandra sphenanthera. Phytochemistry. 2008;69:2862–2868. doi: 10.1016/j.phytochem.2008.09.010. [DOI] [PubMed] [Google Scholar]
- Xiao WL, Wang RR, Zhao W, Tian RR, Shang SZ, Yang LM, Yang JH, Pu JX, Zheng YT, Sun HD. Anti-HIV-1 activity of lignans from the fruits of Schisandra rubriflora. Arch Pharm Res. 2010;33:697–701. doi: 10.1007/s12272-010-0508-7. [DOI] [PubMed] [Google Scholar]
- Xiao WL, Yang LM, Zhang HB, Xue YB, Yang GY, Pu JX, Wang RR, Zheng YT, Sun HD. Chemical constituents from the leaves and stems of Schisandra lancifolia. Chem Pharm Bull. 2010;58:852–855. doi: 10.1248/cpb.58.852. [DOI] [PubMed] [Google Scholar]
- Xue JJ, Cao JL, Yang GY, Pu JX, Sun HD, Hu QF, Xiao WL. Lignans from Schisandra lancifolia. J Asian Nat Prod Res. 2011;13:492–497. doi: 10.1080/10286020.2011.570949. [DOI] [PubMed] [Google Scholar]
- Yang F, Zhang HH, Zhang YY, Chen WS, Yuan HL, Lin HW. A Hepatitis B virus inhibitory neolignan from Herpetospermum caudigerum. Chem Pharm Bull. 2010;58:402–404. doi: 10.1248/cpb.58.402. [DOI] [PubMed] [Google Scholar]
- Yang GY, Fan P, Wang RR, Cao JL, Xiao WL, Yang LM, Pu JX, Zheng YT, Sun HD. Dibenzocyclooctadiene lignans from Schisandra lancifolia and their anti-human Immunodeficiency virus-1 activities. Chem Pharm Bull. 2010;58:734–737. doi: 10.1248/cpb.58.734. [DOI] [PubMed] [Google Scholar]
- Yang GY, Li YK, Wang RR, Li XN, Xiao WL, Yang LM, Pu JX, Zheng YT, Sun HD. Dibenzocyclooctadiene lignans from Schisandra wilsoniana and their anti-HIV-1 activities. J Nat Prod. 2010;73:915–919. doi: 10.1021/np100067w. [DOI] [PubMed] [Google Scholar]
- Yang GY, Li YK, Wang RR, Xiao WL, Yang LM, Pu JX, Zheng YT, Sun HD. Dibenzocyclooctadiene lignans from the fruits of Schisandra wilsoniana and their anti-HIV-1 activities. J Asian Nat Prod Res. 2010;12:470–476. doi: 10.1080/10286020.2010.489823. [DOI] [PubMed] [Google Scholar]
- Yang ZF, Wang YT, Zheng ZG, Zhao SS, Zhao J, Lin Q, Li CY, Zhu Q, Zhong NS. Antiviral activity of Isatis indigotica root-derived clemastanin B against human and avian influenza A and B viruses in vitro. Int J Mol Med. 2013;31:867–873. doi: 10.3892/ijmm.2013.1274. [DOI] [PubMed] [Google Scholar]
- Yang GY, Wang RR, Mu HX, Li YK, Li XN, Yang LM, Zheng YT, Xiao WL, Sun HD. Dibenzocyclooctadiene lignans and norlignans from fruits of Schisandra wilsoniana. J Nat Prod. 2013;76:250–255. doi: 10.1021/np300745q. [DOI] [PubMed] [Google Scholar]
- Yeo H, Li Y, Fu L, Zhu JL, Gullen EA, Dutschman GE, Lee Y, Chung R, Huang ES, Austin DJ, Cheng YC. Synthesis and antiviral activity of helioxanthin analogues. J Med Chem. 2005;48:534–546. doi: 10.1021/jm034265a. [DOI] [PubMed] [Google Scholar]
- Ying CX, Li Y, Leung CH, Robek MD, Cheng YC. Unique antiviral mechanism discovered in anti-hepatitis B virus research with a natural product analogue. Proc Natl Acad Sci USA. 2007;104:8256–8231. doi: 10.1073/pnas.0609883104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ying CX, Tan SL, Cheng YC. Helioxanthin analogue 8–1 inhibits duck hepatitis B virus replication in cell culture. Antivir Chem Chemoth. 2010;21:97–103. doi: 10.3851/IMP1686. [DOI] [PubMed] [Google Scholar]
- Yu JQ, Hang W, Duan WJ, Wang X, Wang DJ, Qin XM. Two new anti-HBV lignans from Herpetospermum caudigerum. Phytochem Lett. 2014;10:230–234. doi: 10.1016/j.phytol.2014.10.001. [DOI] [Google Scholar]
- Yuan HL, Liu Y, Zhao YL, Xiao XH. Herpetin, a new bioactive lignan isolated from Herpetospermum caudigerum. J Chin Pharm Sci. 2005;14:140–143. [Google Scholar]
- Yuan HL, Yang M, Li XY, You RH, Liu Y, Zhu J, Xie H, Xiao XH. Hepatitis B virus inhibiting constituents from Herpetospermum caudigerum. Chem Pharm Bull. 2006;54:1592–1594. doi: 10.1248/cpb.54.1592. [DOI] [PubMed] [Google Scholar]
- Zhang GL, Li N, Wang YH, Zheng YT, Zhang Z, Wang MW. Bioactive lignans from Peperomia heyneana. J Nat Prod. 2007;70:662–664. doi: 10.1021/np0605236. [DOI] [PubMed] [Google Scholar]
- Zhang XJ, Yang GY, Wang RR, Pu JX, Sun HD, Xiao WL, Zheng YT. 7,8-Secolignans from Schisandra wilsoniana and their anti-HIV-1 activities. Chem Biodivers. 2010;7:2692–2701. doi: 10.1002/cbdv.200900367. [DOI] [PubMed] [Google Scholar]
- Zhang HJ, Li WF, Fong HHS, Soejarto DD. Discovery of bioactive compounds by the UIC-ICBG drug discovery program in the 18 years since 1998. Molecules. 2016;21:1448. doi: 10.3390/molecules21111448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang HJ, Rumschlag-Booms E, Guan YF, Liu KL, Wang DY, Li WF, Nguyen VH, Cuong NM, Soejarto DD, Fong HHS, Rong LJ. Anti-HIV diphyllin glycosides from Justicia gendarussa. Phytochemistry. 2017;136:94–100. doi: 10.1016/j.phytochem.2017.01.005. [DOI] [PubMed] [Google Scholar]
- Zhang HJ, Rumschlag-Booms E, Guan YF, Wang DY, Liu KL, Li WF, Nguyen VH, Cuong NM, Soejarto DD, Fong HHS, Rong LJ. Potent inhibitor of drug-resistant HIV-1 strain identified from the medicinal plant Justicia gendarussa. J Nat Prod. 2017;80:1798–1807. doi: 10.1021/acs.jnatprod.7b00004. [DOI] [PubMed] [Google Scholar]
- Zhao JQ, Wang YM, Zhu HT, Wang D, Li SH, Cheng RR, Yang CR, Wang YF, Xu M, Zhang YJ. Highly Oxygenated limonoids and lignans from Phyllanthus flexuosus. Nat Prod Bioprospect. 2014;4:233–242. doi: 10.1007/s13659-014-0026-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu YK, Guan J, Xiao ZY, Cosentino LM, Lee KH. Anti-AIDS agents. Part 61: Anti-HIV activity of new podophyllotoxin derivatives. Bioorgan Med Chem. 2004;12:4267–4273. doi: 10.1016/j.bmc.2004.04.048. [DOI] [PubMed] [Google Scholar]
- Zinser E, Krawczyk A, Muhl-Zurbes P, Aufderhorst U, Drabner C, Stich L, Zaja M, Strobl S, Steinkasserer A, Heilingloh CS. A new promising candidate to overcome drug resistant herpes simplex virus infections. Antivir Res. 2018;149:202–210. doi: 10.1016/j.antiviral.2017.11.012. [DOI] [PubMed] [Google Scholar]