Abstract
Multidrug resistance (MDR) is one of the main impediments in the treatment of cancers. MDR cancer cells are resistant to multiple anticancer drugs. One of the major mechanisms of MDR is the efflux of anticancer drugs by ABC transporters. Increased activity and overexpression of these transporters are important causes of drug efflux and, therefore, resistance to cancer chemotherapy. Overcoming MDR is a fundamental prerequisite for developing an efficient treatment of cancer. To date, various types of ABC transporter inhibitors have been employed but no effective anticancer drug is available at present, which can completely overcome MDR. Phytochemicals can reverse MDR in cancer cells via affecting the expression or activity of ABC transporters, and also through exerting synergistic interactions with anticancer drugs by addressing additional molecular targets. We have listed numerous phytochemicals which can affect the expression and activity of ABC transporters in MDR cancer cell lines. Phytochemicals in the groups of flavonoids, alkaloids, terpenes, carotenoids, stilbenoids, lignans, polyketides, and curcuminoids have been examined for MDR-reversing activity. The use of MDR-reversing phytochemicals with low toxicity to human in combination with effective anticancer agents may result in successful treatment of chemotherapy-resistant cancer. In this review, we summarize and discuss published evidence for natural products with MDR modulation abilities.
Keywords: cancer, ABC-transporter, drug efflux, multidrug resistance, secondary metabolites, synergism
Introduction
Besides surgery and radiation, chemotherapy is one of the standard treatments of cancer. Drugs used in chemotherapy usually disturb cell division by inhibition of microtubule formation or disassembly (vinca alkaloids, paclitaxel), DNA topoisomerase (camptothecin and derivatives) or they intercalate or alkylate DNA (doxorubicin, cisplatin) (Wink, 2007; Wink et al., 2012). Furthermore, chemotherapy often causes extreme side effects as these drugs also affect the division of normal cells or they cause mutations, which can lead to secondary cancers. This additionally leads to restriction of the therapeutic applications; both dosage and application interval must be kept limited. When chemotherapeutic agents are used, it is often a matter of time before the cancer cells develop resistance against them. One of the major resistance mechanisms is the overexpression of ABC transporters, which can pump out the chemotherapeutic from cancer cells. Because these ABC transporters have a wide substrate spectrum, they not only confer resistance to a single drug but to several others, therefore, the term Multidrug Resistance (MDR). Since multiple drug resistance is a major issue in tumor therapy, new strategies are necessary to overcome this obstacle. One strategy involves the combination of anti-cancer drugs with modulators of ABC transporters. Our review presents a summary about various modulating effects of phytochemicals.
Multidrug Resistance in Cancer
One of the major difficulties in suppressing growth and survival of cancer cells is multidrug resistance. MDR is the resistance of cancer cells to various types of anticancer drugs which may have an intrinsic or acquired origin. Acquired resistance is induced after the administration of chemotherapy, whereas, intrinsic resistance already exists prior to drug application in cancer cells (Wang et al., 2019). Several mechanisms in cancer cells can lead to MDR (Coley, 2008). These include changes in target enzymes, such as DNA topoisomerases (Brown et al., 1995), alteration in microtubule-associated proteins (Zhang et al., 1998), mutations or changes in tubulin (Kamath et al., 2005; Wang and Cabral, 2005), alteration in microtubules (Kavallaris et al., 2001), mitotic arrest (Kamath et al., 2005), mutated protein p53 (O'Connor et al., 1997), disruption in DNA repair due to the damaging effect of an anticancer drug (Bernstein et al., 2002) and the impairment of apoptosis or genes involved in apoptosis and necrosis (Tanaka et al., 2000; Simstein et al., 2003).
A widespread mechanism of MDR is drug efflux via transmembrane transporters known as ATP-binding cassette transporters (ABC transporter). Overexpression of these transporters is the most important cause of drug resistance in many cancer cells. The family of ABC transporter proteins has 48 members in humans and far more in nature (Chen et al., 2016). The most well-known and widely-studied ABC transporters include P-Glycoprotein (P-gp), multidrug resistance protein 1 (MRP1), and breast cancer resistance protein (BCRP). These transporters are expressed in healthy cells of various mammalian tissues having physiological tasks for translocating small molecules. They are found especially in intestinal epithelial cells, endothelial cells of blood capillaries and epithelia of renal proximal tubules being involved in the excretion and clearance of endogenous and exogenous cytotoxic substances (Borst et al., 2000; Durmus et al., 2015; Chen et al., 2016). ABC transporters evolved in nature millions of years ago to eliminate toxic phytochemicals that herbivores would obtain from their plant diet (Wink, 2007). Anticancer drugs also can be substrates of these transporters and if being considered as foreign they get exported to the extracellular space by cells expressing ABC transporters. Cancer cells which are not resistant to anticancer agents yet can develop the ability by overexpressing these transporters for saving themselves from the substances being toxic to them. This leads to an increased efflux, leading to low intracellular drug concentrations, insufficient to kill a cancer cell. Once this overexpression has occurred, the efflux also affects other chemotherapeutics and thus, makes the cancer cell resistant to chemotherapy (Coley, 2008; Durmus et al., 2015).
P-gp (ABCB1)
P-gp is a 170 kDa protein which is encoded by the MDR1 gene. This transporter is found in normal cells of various tissues including the brain, liver, kidney, gastrointestinal tract and pancreas. P-gp transports anticancer drugs such as paclitaxel, doxorubicin, daunorubicin, epirubicin, mitoxantrone, vincristine, and vinblastine against the concentration gradient using energy derived from hydrolysis of ATP (Chen et al., 2016). Chemotherapeutic agents can stimulate P-gp expression in cancer cells and thereby cause resistance to chemotherapy. Chemotherapy has been reported to increase the proportion of P-gp-expressing tumors by approximately 1.8-fold in breast cancer. Moreover, in patients with activated P-gp transporter in their tumors, the risk of failure of chemotherapy is 3 times higher than in patients who do not express P-gp transporter (Trock et al., 1997).
Multidrug Resistance Proteins (MRPs)
Another class of membrane transporters which causes MDR is MRPs. Nine members of this class have been identified so far (König et al., 2005; Coley, 2008). MRPs are found in normal cells of some mammalian tissues and expel drugs as a complex with glutathione, glucuronate, or sulfate (Borst et al., 2000; Coley, 2008). Among the MRP transporters, MRP1 (ABCC1) is the most important and most studied one regarding MDR. The MRP1 protein has a molecular weight of 190 kDa. Similar to P-gp, MRP1 expression has been reported to be considerably higher expressed in cancer cells after chemotherapy than before chemotherapy (Trock et al., 1997). Therefore, MRP1 enhances resistance to chemotherapy and to anticancer drugs such as doxorubicin, daunorubicin, epirubicin, vincristine, and vinblastine (Coley, 2008).
BCRP (ABCG2)
Breast cancer resistance protein, also called mitoxantrone transporter (MXR1), has a molecular weight of 72 kDa. BCRP is extensively expressed in MCF-7 breast cancer cells (Doyle et al., 1998). This protein is also expressed in other tissues including the liver, kidney, and intestine (Chen et al., 2016). The anticancer drugs doxorubicin, daunorubicin, epirubicin and mitoxantrone have been described as substrates of BCRP transporter (Coley, 2008). Thus, cancer cells overexpressing BCRP transporter become resistant to these drugs.
MDR Modulators
One of the essential requirements for developing better anti-cancer therapies is overcoming multidrug resistance. Much research has been carried out on cancer treatment and development of anticancer drugs in recent years but MDR to cytostatics is still a great impediment. Although our knowledge about the mechanisms of multidrug resistance has increased, there is no effective drug which can completely overcome or reverse resistance at non-toxic concentrations. Since ABC transporters play a fundamental role in resistance to chemotherapy, the ability to inhibit them in a combination with conventional treatments will greatly help to treat cancer (Chen et al., 2016).
Until now, different types of ABC transporter inhibitors have been examined. The use of the first generation of these compounds, including verapamil and cyclosporine A, in combination with anticancer drugs had poor clinical success and toxic effects (Daenen et al., 2004). Second generation of MDR modulators included dexverapamil, valspodar, and dexniguldipine. Even though less toxic and with a higher therapeutic index than the first generation, this group of modulators is not well suited for a therapy either, both because of its interactions with other drugs and ABC transporters, as well as due to the inhibition of enzymes like CYP3A (Wandel et al., 1999; Syed and Coumar, 2016). The third-generation ABC transporter modulators do not have the disadvantages of the first and second generation. They are potent and non-competitive inhibitors of P-gp, and also less toxic. Tariquidar (XR9576) and zosuquidar are members of the third generation of MDR modulators but unfortunately they were not efficative in clinical trials (Cripe et al., 2010; Kelly et al., 2011).
Phytochemicals
Alkaloids (Figure 1) are the most widely studied group of secondary metabolites in terms of MDR, not only because of their quantity but also because of their great diversity (Wink, 2007; Wink et al., 2012). As alkaloids have a wide distribution among angiosperms (Wink, 2020) and represent a diversity of structures, they differ in pharmacological and toxicological properties. Alkaloids contain heterocyclic nitrogen, which mostly has its origin in amino acids (Cseke et al., 2006). Alkaloids are subdivided into many subcategories of special functional groups, similarities of skeleton or biosynthetic pathways.
Figure 1.
Chemical structures of some selected alkaloids with MDR reversal effects.
Quinoline and isoquinoline alkaloids for example have a benzopyridine ring differing in the position of their nitrogen. Quinazoline alkaloids have a similar aromatic structure but with two nitrogen atoms instead of one. Each of these structures has found several uses depending on molecular structure. There are many examples for quinoline alkaloids such as mefloquine as antimalarial agent, fluoroquinolone antibiotics and topotecan as anticancer drug, just to name a few (Collin and Höke, 2000; Tiwary et al., 2015). The latter two work as inhibitors of different DNA topoisomerases (Lemmer and Brune, 2004).
Quinolizidines also are cyclic nitrogen-containing compounds but unlike the previously mentioned subgroups they are not aromatic. A natural representative is sparteine, which is used as an antiarrhythmic agent blocking sodium channels (Ruenitz and Mokler, 1977; Körper et al., 1998; Gawali et al., 2017).
Among other groups of alkaloids, indole, monoterpene indole and β-carboline alkaloids show many pharmacological activities (Gilbert, 2001). A typical basic structure of β-carboline alkaloids consists of benzene fused with a five-membered pyrrole and is consequently similarly structured to some endogenous hormones and neurotransmitters such as serotonin and melatonin. In addition to benzene, the pyrrole ring of β-carboline is fused to a pyridine, another six-membered nitrogen-containing ring. This structure by itself is an inverse agonist of GABA-receptors, which involves psychological influence on humans (Aktories et al., 2017). Substrates among the indoles target many receptors, for example PDE-receptors e.g. by tadalafil, 5-HT receptors e.g. by naratriptane and HMG-CoA reductases e.g. by fluvastatin (Wink, 2000; de Sa et al., 2009). Several indole alkaloids have stimulant and hallucinogenic properties (Wink, 2000; Wink and van Wyk, 2008).
As the nitrogen of steroidal alkaloids does not originate from amino acids they belong to pseudoalkaloids. A member of steroidal alkaloids is the teratogenic cyclopamine, which can cause cyclopean eyes in vertebrates (Roberts and Wink, 1998; Incardona et al., 1998).
Piperidine and diketopiperazine are six-membered non-aromatic moieties in alkaloids whereby diketopiperazine is a cyclic dipeptide having two oppositely located nitrogen. Pyrazine has the same position of nitrogen, though aromatic. Here we also can find medicinal use of active ingredients such as the oxytocin antagonist retosiban and plinabulin which is still in clinical trial against multiple drug resistant non-small cell lung cancer (Borthwick and Liddle, 2011; Mohanlal et al., 2019).
Tropane alkaloids are widespread and their plants one of the oldest medicines to use because of spasmolytic, mydriatic and hallucinogenic properties (Wink and van Wyk, 2008; van Wyk and Wink, 2017). They contain a special bicyclic moiety which is made of a seven-membered ring and a nitrogen atom which is linked to its C-1 and C-5 and forms the second ring (Osman et al., 2013). Due to their spasmolytic effect nowadays we use tropane alkaloids such as scopolamine or atropine for digestive tract spastic conditions and for ophthalmological purposes (Kukula-Koch and Widelski, 2017).
The most popular alkaloid caffeine belongs to purine alkaloids, which are consumed by many people on all continents. Theobromine, theophylline and caffeine are common members found as main ingredients in chocolate, mate, cola, green and black tea or coffee (Baumann and Frischknecht, 1988). By inhibiting adenosine receptors and cAMP phosphodiesterase they can mediate a stimulant effect (van Wyk and Wink, 2017).
Flavonoids (Figure 2) are another complex but also often colored group of secondary metabolites. Unlike alkaloids, we can make a general statement about their common origin and basic skeleton. Flavonoids can be classified as polyphenols which share a common biosynthesis. They contain aromatic rings with phenolic hydroxyl groups. These phenolic hydroxyl groups can dissociate under physiological condition and form negatively charged phenolate ions. Because of these properties flavonoids and polyphenols can interact with proteins forming multiple hydrogen and ionic bonds (Wink, 2015). Flavonoids are widely distributed in plants and are responsible, inter alia, for their pollinator attracting colors, ultraviolet light protection, antioxidant and antimicrobial functions, and mediating symbiosis with bacteria. Including the main subgroups flavones, isoflavones, flavonols, flavanones, anthocyanins, chalcones and catechins, they derive from flavan, a benzopyran structure with a phenyl ring in position 2. Flavonoids with a phenyl ring in position 3 and 4 are called iso- and neoflavonoids, respectively. The variety of flavonoids comes from many functional groups and different states of oxidation of the heterocycle (Koes et al., 1994; Wink and van Wyk, 2008; Hänsel and Sticher, 2009). Although there has been a lot of research on the antioxidant capacity of flavonoids, the mechanism is not fully understood yet. Many studies have reported anti-inflammatory, anti-carcinogenic, anti-mutagenic, antiviral, anti-allergic and osteogenetic potentials of flavonoids in vitro. There is evidence, that polyphenols are also important for the pharmacological activity of many medicinal plants (van Wyk and Wink, 2017). Still there is a lack of information about how the necessary bioavailability is achieved in the human body as polyphenols are polar compounds (Panche et al., 2016). Most common representatives in food are luteolin and apigenin. Isoflavones are known for their estrogenic properties (Wink, 2015).
Figure 2.
Chemical structures of some selected flavonoids with MDR reversal effects.
In addition to flavonoids, there are smaller but also important groups of polyphenols, for example stilbenoid with resveratrol as its main member known for its potential as anti-cancer (Huang et al., 2014), antioxidant and anti-aging agent (Alamolhodaei et al., 2017). Curcuminoids (Figure 3) have been widely studied and have been found to have many functions such as antioxidative, anti-cancer, anti-microbial and anti-inflammatory effects in humans. They can interact with many targets (Fantini et al., 2015).
Figure 3.
Chemical structures of some selected curcuminoids with MDR reversal effects.
Terpenes (Figure 4) are widely distributed in plants, fungi and animals. They are composed of different numbers of isoprene units, forming mono- (C10), di- (C20), sesqui- (C15), tetra- (C40) and triterpenes (C30). Many mono- and sequiterpenes are volatile and aromatic and typical ingredient of essential oils. These compounds are often lipophilic and can thus modulate the fluidity and permeability of biomembranes in animals and microbes. Many plants with essential oil have been used in traditional medicine for treatment of microbial infections and inflammation (Wink, 2015; van Wyk and Wink, 2017).
Figure 4.
Chemical structures of some selected terpenes with MDR reversal effects.
Known for their skin permeation enhancing ability, terpenes have been used as moieties of synthetic structures for topical use (Smith and Maibach, 1995). In vitro studies have shown anti-cancer, antimicrobial and antioxidant activities but for practice and use in humans this class of secondary metabolites must be further investigated (Lu et al., 2012; Cör et al., 2018). Squalene is a standard triterpene produced in plants and animals, and it is the precursor for steroid synthesis. Glycosides of steroids or triterpenes, so-called saponins have one or more polysaccharides attached saponins are amphiphilic and react as a detergent. They generally form stable foams and complex cholesterol in biomembranes. As a consequence, saponins can lyse biomembranes (Bloch, 1983). In traditional medicine they have been used for example as expectorants and anti-infectants (Wink, 2015). As effective components of vaccine adjuvants they enhance the cellular immune response (Sun et al., 2009).
Carotenoids are tetraterpenes with many conjugated double bonds. They often exhibit yellow to purple colors and can function as anti-oxidants and precursors for vitamin A.
As the name implies polyketides contain carbonyl groups positioned between methylene groups. Still they vary in shape and volume. Although they appear with an impressive structural variety, they have their source from the same biosynthetic pathway. The most famous members among drugs may be the antibiotic erythromycin or the antifungal amphotericin B.
Phytochemicals Modulating MDR Targeting ABC-Transporters
A considerable number of secondary metabolites, which affect ABC transporters has already been discovered (Table 1) and several of them will be discussed in the following; glaucine (an isoquinoline alkaloid) increased the efflux of substrates such as ADR and MTX in the P-gp over-expressing cell lines of MCF-7/ADR and reduced DOX resistance in Caco-2 and CEM/ADR5000 (Eid et al., 2013; Lei et al., 2013). Tetrandrine (a benzolisoquinoline alkaloid) also caused an inhibition of efflux in Caco-2 and CEM/ADR5000 cells (Sun and Wink, 2014). A 5-substituted derivative of it named PY35 was tested for the MDR reversal activity, and showed more MDR reversal than the natural compound in resistant K562/ADM and MCF-7/ADM cells (Cao et al., 2014). Hernandezine, a bisbenzyl-isoquinoline, is a potent inhibitor of P-gp in MDR19-HEK293 cells and was able to resensitize MDR19-HEK293 and KB-V-1 cells to DOX after entering the cell membrane (Hsiao et al., 2016). High MDR-reversing activities of the quinoline derivatives were linked to the presence of two aryl rings in the hydrophobic moiety, deviation of the aryl rings from a common plane, basicity of nitrogen atom in piperazine, as well as to the distance between the hydrophobic moiety and the basic nitrogen of piperazine which must be no less than 5 Å (Suzuki et al., 1997). The quinolyl group was also suggested to have a key role in the activity of quinolines, because substitution of a quinoline ring by a naphthyl ring or a phenyl ring resulted in the reduction of MDR-reversing activity of the compounds (Suzuki et al., 1997). Among indole alkaloids, antofine showed synergistic effects with PTX in A549-PA cells and overcame resistance to PTX (Kim et al., 2012). The β-carboline harmine reversed the resistance of MTX and CPT in MDA-MB-231 cells with BCRP as overexpressed transporter but it could not affect the P-gp over-expressing CEM/ADR5000 cells the same way in this study (Ma and Wink, 2010). Not only has harmine been tested in combination with DOX but also as three-drug-combination with DTN both of which showed here an increase of effect on cells and a reduction of DOX resistance in Caco-2 and CEM/ADR5000 cells (Eid et al., 2013). As an N-acylpiperidine, piperine inhibited the efflux of the tested substrate which led to an increase of its concentration in Caco-2 and CEM/ADR5000 cells (Li et al., 2018a). Tests of piperine on MCF-7/DOX and A-549/DDP cells resulted in an increase in cytotoxicity of MTX and DOX (Li et al., 2011). Gravacridonetriol, gravacridonediol and its monomethyl ether, all had an inhibitive effect on P-gp L5178/MDR1 cells, which also led to a higher cytotoxicity of DOX (Rethy et al., 2008). Substitution of methyl groups at the positions C-2 and C-4 of acridone led to the increased lipophilicity, which can enhance the binding affinity of acridone derivatives to P-gp (Mayur, 2015). Murahari et al. (2017) studied on 2,4-dimethylacridone derivatives and showed that these compounds were potential modulators of P-gp-mediated MDR. 2,4-dimethylacridones are tricyclic and hydrophobic and have methyl groups at C-2 and C-4 positions with a propyl or butyl side chain containing terminally substituted tertiary amino groups. They found that an alkyl side chain and hydroxyl substituted secondary amine are necessary for the acridones to reverse the P-gp-mediated multidrug resistance. Moreover, an alkyl side chain of length four (butyl) was found to have higher biological activity (Murahari et al., 2017). Hegde et al. (2004) reported that replacing the hydrogen atom at position C-4 by a methoxy group slightly enhanced the lipophilicity of the acridone derivatives. Furthermore, they investigated the MDR-reversing activity of N-10-substituted acridones and N-10-substituted 4-methoxyacridones relative to their corresponding unsubstituted counterparts where the C-4 positions of the acridone rings are occupied by a hydrogen atom and a methoxy group, respectively. The parent acridone and 4-methoxyacridone had the least effect in inhibiting drug efflux suggesting that N-10-substitution is necessary for an ideal activity. It was also found that 4-methoxyacridone derivatives are more efficient than their acridone derivatives' counterparts in increasing drug accumulation. Several N-10-substituted acridones and N-10-substituted-4-methoxyacridones showed MDR-reversing activity greater than the P-gp inhibitor verapamil (Hegde et al., 2004). In another study eighteen N-10-substituted-2-bromoacridones were examined for the anti-MDR activity and compared to the parent compound 2-bromo-10H-acridin-9-one. N-10-substitution was suggested to be necessary for optimal activity of 2-bromoacridones because the parent compound had the least effect in efflux inhibiting activity (Mayur et al., 2006).
Table 1.
Phytochemicals modulating MDR via ABC-transporters.
The effects of secondary metabolites on different cell lines expressing ABC-transporters – Transporters targeted directly | |||||
---|---|---|---|---|---|
Substance | Cell line | Assay system | Result | Reference | |
Alkaloids | |||||
Quinolines, Isoquinolines, Quinazolines | Dauriporphine | MES-SA/DX5 and HCT15 | MDR reversing activity (cytotoxicity assay in the presence and absence of an anticancer drug, PTX) | Increase in cytotoxicity of PTX (inhibition of P-gp MDR) | Min et al. (2006) |
Fangchinoline | MDR1-MDCK II | MDR reversing activity | Decrease in substrate (PTX) efflux, inhibition of the multidrug resistance of antitumor drug PTX | He et al. (2010) | |
Glaucine | Caco-2, CEM/ADR5000 | MTT assay, Rh-123 accumulation assay using fluorospectroscopy | Reversal of DOX resistance in both cell lines with very high effect, synergism with DOX | Eid et al. (2013) | |
Caco-2, CEM/ADR5000 | MTT assay, Rh-123 accumulation assay using fluorospectroscopy | Sensitization of cell lines, enhancement of cytotoxicity, strong reduction of the IC50 value of DOX and consequently increase of efficacy, synergism with DOX and DTN | Eid et al. (2013) | ||
MCF-7/ADR | MDR reversing activity, ADR and MTX efflux assay, real-time RT-PCR, P-gp and MRP1 ATPase activity assay | Inhibition of P-gp and MRP1-mediated efflux, suppression of the expression of MDR1 and MRP1 genes, reversion of the resistance of MCF-7/ADR to ADR and MTX, increase in P-gp and MRP1 ATPase activities | Lei et al. (2013) | ||
Hernandezine | MDR19-HEK293, KB-V-1, NCI-ADR-RES |
MDR reversing activity, fluorescent drug (calcein-AM and pheophorbide A) accumulation assay, | Inhibition of the transport function of ABCB1 (P-gp), increase in calcein-AM accumulation in MDR19-HEK293 cells, resensitizing of MDR19-HEK293 cells to DOX, resensitizing of KB-V-1 cells to DOX, colchicine and VCR, resensitizing of NCI-ADR-RES cells to DOX, colchicine and vincristine | Hsiao et al. (2016) | |
Roemerine | MDR KB-V1 | Cytotoxicity assay with VBL | Cytotoxicity synergism | You et al. (1995) | |
Sanguinarine | Caco-2, CEM/ADR5000 | MTT assay, Rh-123 accumulation assay using fluorospectroscopy | Reversal of DOX resistance in both cell lines with very high effect, synergism with DOX | Eid et al. (2013) | |
Caco-2, CEM/ADR5000 | MTT assay, Rh-123 accumulation assay using fluorospectroscopy | Sensitization of cell lines to DOX | Eid et al. (2013) | ||
Tetrandrine | Caco-2, CEM/ADR5000 | Rh-123 accumulation | Increased accumulation of substrate, inhibited efflux in both cell lines, reduction of P-gp expression | Sun and Wink (2014) | |
Quinidine homodimer | MCF-7/DX1 | Radioactive substrate ([3H]PTX) accumulation assay, flow cytometric accumulation assay, confocal microscopy | Inhibition of the efflux of Rh-123, DOX, BODIPY-FL-prazosin, PTX and MTX, | Pires et al. (2009) | |
Steroidal alkaloids | Verabenzoamine | L5178/MDR1 (human MDR1-gene-transfected mouse lymphoma cells) | Flow cytometric assay of Rh-123 accumulation | Increase in accumulation | Ivanova et al. (2011) |
Veralosine | L5178/MDR1 | Flow cytometric assay of Rh-123 accumulation | Increase in accumulation | Ivanova et al. (2011) | |
Veralosinine + Veranigrine | L5178/MDR1 | Flow cytometric assay of Rh-123 accumulation | Increase in accumulation | Ivanova et al. (2011) | |
Indoles and β-carbolines | Antofine | A549-PA (PTX-resistant human lung cancer cell line) | P-gp expression using western blot, MDR-1 mRNA expression using RT-PCR, Rh-123 accumulation by FACS | Reduction of P-gp and MDR-1 mRNA expression, increase in intracellular Rh-123 content, synergism with PTX | Kim et al. (2012) |
Arboloscine | KB/VJ300 | MDR reversing activity | Moderate to weak activity in reversing MDR | Gan et al. (2014) | |
Conoduramine | KB-V1 | Binding assay (VBL binding to KB-V1 vesicles) | Inhibition of drug-binding, resulting in circumventing multi-drug resistance | You et al. (1994) | |
Coronaridine | KB-V1 | Binding assay (VBL binding to KB-V1 vesicles) | Inhibition of drug-binding resulting in circumventing multi-drug resistance | You et al. (1994) | |
Harmine | MDA-MB-231 (BCRP), CEM/ADR5000 (P-gp) | Rh-123 accumulation assay, cytotoxicity assay using MTT, MTX efflux assay | Reversal of MTX and CPT resistance in cell line with BCRP-mediated efflux, no effect on P-gp mediated efflux | Ma and Wink (2010) | |
Caco-2, CEM/ADR5000 | MTT assay | Reversal of DOX resistance in both cell lines, synergism with DOX | Eid et al. (2013) | ||
Caco-2, CEM/ADR5000 | MTT assay, Rh-123 accumulation assay using fluorospectroscopy | Sensitization of cell lines to DOX | Eid et al. (2013) | ||
Kopsamine | KB/VJ300 | MDR reversing activity | Appreciable activity in reversing MDR | Kam et al. (1998) | |
Kopsiflorine | KB/VJ300 | MDR reversing activity | Appreciable activity in reversing MDR | Kam et al. (1998) | |
Lahadinine A | KB/VJ300 | MDR reversing activity | Appreciable activity in reversing MDR | Kam et al. (1998) | |
Leuconicine (types A - E) | KB/VJ300 | MDR reversing activity | Reversal of multidrug resistance | Gan et al. (2009) | |
11-Methoxykopsilongine | KB/VJ300 | MDR reversing activity | Appreciable activity in reversing MDR | Kam et al. (1998) | |
N-methoxycarbonyl-11,12-methylene-dioxykopsinine | KB/VJ300 | MDR reversing activity | Appreciable activity in reversing MDR | Kam et al. (1998) | |
Pleiocarpine | KB/VJ300 | MDR reversing activity | Appreciable activity in reversing MDR | Kam et al. (1998) | |
Reserpine | CEM/VLB100 | MDR reversing activity | Increase in cytotoxicity, synergism with VBL | Pearce et al. (1989) | |
Tryptanthrin | Caco-2 | Transport across the Caco-2 cell monolayers, MDR1 & 2 gene expression | Decrease in the efflux transport of the P-gp and MRP2 substrates (potential inhibitor of P-gp and MRP2) | Zhu et al. (2011) | |
Vocamine | KB-V1 | Binding assay (VBL binding to KB-V1 vesicles) | Inhibition of drug-binding resulting in circumventing multi-drug resistance | You et al. (1994) | |
Yohimbine | CEM/VLB100 | MDR reversing activity | Increase in cytotoxicity synergism with VBL | Pearce et al. (1989) | |
Piperidines, Pyrazines, Diketopiperazines | 11E-didehydrostemofoline, 11Z-didehydrostemofoline | K562/Adr | MTT assay; fluorescent substrates accumulation assays | Increase in sensitivity to DOX and PTX; increase in the intracellular concentrations of Rh-123 and calcein-AM | Umsumarng et al. (2017) |
Isostemofoline | K562/Adr | MTT assay; fluorescent substrates accumulation assays | Increase in sensitivity to DOX and PTX; increase in the intracellular concentrations of Rh-123 and calcein-AM | Umsumarng et al. (2017) | |
Lobeline | Caco-2, CEM/ADR5000 | Rh-123 accumulation assay, cytotoxicity assay using MTT, MTX efflux assay | Inhibition of P-gp mediated efflux, accumulation of Rh-123, increase of DOX sensitivity of both cell lines | Ma and Wink (2008) | |
Oxystemokerrine | KB-V1 | MDR reversing activity | Slight increase in sensitivity | Chanmahasathien et al. (2011) | |
Piperine | MCF-7/DOX, A-549/DDP | MDR reversing activity, Rh-123 accumulation assay, MTX efflux assay, RT-PCR | Reversal of resistance, decrease in ABCB1 and ABCG2 genes expression in MCF-7/DOX cells, decrease in ABCC1 gene expression in A-549/DDP cells | Li et al. (2011) | |
Caco-2, CEM/ADR 5000 | Cytotoxicity assay using MTT, Rh-123 and calcein-AM retention assay | Substrate, synergistic enhancement of cytotoxicity, inhibition of efflux and consequently accumulation of Rh-123 and calcein-AM | Li et al. (2018a) | ||
Stemocurtisine | KB-V1 | MDR reversing activity | Slight increase of sensitivity | Chanmahasathien et al. (2011) | |
Stemofoline | KB-V1 | MDR reversing activity | Increase in cell sensitivity | Chanmahasathien et al. (2011) | |
Tetramethylpyrazine | MCF-7/Dox | Flow cytometric evaluation of DOX accumulation, P-gp expression | Inhibition of efflux, decrease in P-gp expression | Zhang et al. (2012) | |
Tropane alkaloids | Pervilleine A | MDR KB-V1 | Intracellular VBL accumulation, cytotoxicity | Cytotoxicity synergism with VBL, increase in VBL accumulation | Mi et al. (2001) |
Pervilleine B, C | MDR KB-V1 | Cytotoxicity assay | Cytotoxicity synergism with VBL | Mi et al. (2002) | |
Acridone alkaloids | Acrimarine E | K562/R7 | DNR accumulation assay | Inhibition of P-gp-mediated drug efflux | Bayet et al. (2007) |
Gravacridonediol | L5178/MDR1 | Rh-123 accumulation assay, MTT assay, MDR1 mRNA expression | Increase in Rh-123 accumulation, increase in DOX toxicity (synergism) | Rethy et al. (2008) | |
Gravacridonediol monomethyl ether | L5178/MDR1 | Rh-123 accumulation assay, MTT assay, MDR1 mRNA expression | Increase in Rh-123 accumulation, cytotoxicity, synergism with DOX, decrease in MDR1 mRNA | Rethy et al. (2008) | |
Gravacridonetriol | L5178/MDR1 | Rh-123 accumulation assay, MTT assay, MDR1 mRNA expression | Increase in Rh-123 accumulation, cytotoxicity, synergism with DOX, decrease in MDR1 mRNA | Rethy et al. (2008) | |
2-Methoxycitpressine I | K562/R7 | DNR accumulation assay | Inhibition of p-glycoprotein-mediated drug efflux | Bayet et al. (2007) | |
Rutacridone | L5178/MDR1 | Rh-123 accumulation assay, MTT assay, MDR1 mRNA expression | Increase in Rh-123 accumulation | Rethy et al. (2008) | |
Purine alkaloids | Olomoucine II | MDCKII-ABCB1, HCT-8 and HepG2 | Hoechst 33342 and DNR accumulation assay | Synergism with DNR (increase in intracellular retention of DNR) | Cihalova et al. (2013} |
Purvalanol A | MDCKII-ABCB1, HCT-8 and HepG2 | Hoechst 33342 and DNR accumulation assay | Synergism with DNR (increase in intracellular retention of DNR) | Cihalova et al. (2013) | |
Roscovitine | MDCKII-ABCB1, HCT-8 and HepG2 | Hoechst 33342 and DNR accumulation assay | Synergism with DNR (increase in intracellular retention of DNR) | Cihalova et al. (2013) | |
Further alkaloids | Anandamine | HK-2 | Fluorimetric measurement of the intracellular accumulation of calcein | Inhibition efflux (increase in the intracellular accumulation of calcein) | Nieri et al. (2006) |
Capsaicin | KB-C2 | Determination of DNR and Rh-123 accumulation | Increase in the accumulation of DNR and Rh-123 | Nabekura et al. (2005) | |
Caco-2 | [3H]-digoxin transport assay | At non-cytotoxic concentrations, inhibition of P-gp mediated efflux transport of [3H]-digoxin | Han et al. (2006) | ||
Caco-2, CEM/ADR 5000 | Cytotoxicity assay using MTT, Rh-123 and calcein-AM retention assay | Substrate, synergistic enhancement of cytotoxicity, inhibition of efflux and consequently accumulation of Rh-123 and calcein-AM | Li et al. (2018a) | ||
Galantamine dimer | MCF-7/DX1 | Measuring Rh-123 and DOX accumulation | Inhibition of efflux (increased accumulation) | Namanja et al. (2009) | |
Polyphenols | |||||
Flavonoids | Acacetin | K562/BCRP | Cytotoxicity assay | Increase in cytotoxicity of SN-38 and MTX, strong reversing activity of BCRP-mediated drug resistances | Imai et al. (2004) |
MDA-MB-231 | BCECF accumulation assay | Inhibition of the efflux of MRP1 fluorescent substrate (BCECF) from breast cancer cells | Wesołowska et al. (2009) | ||
Afrormosin | L5178/MDR1 | Cytotoxicity assay, Rh-123 accumulation assay | Moderately effective on the human MDR1-transfected mouse lymphoma cell line | Gyémánt et al. (2005) | |
MDA-MB-231 | Cytotoxicity assay, BCECF-AM accumulation assay | MRP-mediated efflux pump modifiers, additive effect in combination with epirubicin | Gyémánt et al. (2005) | ||
Amorphigenin | L5178/MDR1 | Cytotoxicity assay, Rh-123 accumulation assay | P-gp-mediated efflux pump modifier (strong MDR-reversal effects), strong antiproliferative effects, synergistic effects in combination with epirubicin | Gyémánt et al. (2005) | |
Apigenin | MCF-7 MX100 | MTX accumulation, cytotoxicity assay | Increasing accumulation and inhibition of BCRP in combination with other flavonoids e.g. biochanin A, and chrysin | Zhang et al. (2004) | |
K562/BCRP | Cytotoxicity assay | Strong reversing activity of BCRP-mediated drug resistances | Imai et al. (2004) | ||
Ampelopsin | K562/ADR | MTT assay, P-gp expression assay using PE-labeled antibody, ADR accumulation assay | Decreasing P-gp expression, reversal of MDR to ADR, increase in cytotoxicity and the intracellular ADR accumulation | Ye et al. (2009) | |
7,8-Benzoflavon | MCF-7 MX100 | Topotecan accumulation studies | Inhibition of the efflux, increasing the accumulation of topotecan | Zhang et al. (2005) | |
MCF-7/ADR | Daunomycin accumulation assay | Decrease of daunomycin efflux, increase in [3H]-daunomycin accumulation, strongly potentiated cytotoxicity of daunomycin | Chung et al. (2005) | ||
Biochanin A | MDA435/LCC6, MCF-7/ADR, MDA435/LCCMDR1 |
Daunomycin accumulation, DOX cytotoxicity | Increase in [3H]-daunomycin accumulation, potentiation of DOX cytotoxicity, inhibition of P-gp-mediated cellular efflux | Zhang and Morris (2003) | |
Panc-1 | Determination of daunomycin and VBL accumulation | Increase in accumulation of daunomycin and VBL in Panc-1 cells, inhibiting MRP1-mediated drug transport | Nguyen et al. (2003) | ||
MCF-7 MX100 | MTX accumulation, cytotoxicity assay | Increasing accumulation and inhibiting BCRP in combination with other flavonoids e.g. apigenin or chrysin | Zhang et al. (2004) | ||
Caco-2 | Ochratoxin A (OTA) accumulation assay | Increase in OTA accumulation, impairing OTA efflux through competitive inhibition of MRP-2 pump | Sergent et al. (2005) | ||
Catechin | Rats jejunal membrane in vitro | P-gp stimulation/inhibition profiles using a P-gp-dependent ATPase assay | Inhibitory effect on P-gp ATPase activity | Najar et al. (2010) | |
NIH-3T3-G185 | Intracellular retention of Rh-123 or LDS-751 (P-gp marker substrate) | Slight facilitation of the efflux of LDS-751 (providing a chemoprotective role) | Wang et al. (2002) | ||
Chalcone | Panc-1 | Determination of daunomycin and VBL accumulation | Increasing the accumulation of daunomycin and VBL in Panc-1 cells, inhibiting MRP1-mediated drug transport | Nguyen et al. (2003) | |
Chrysin | CaCo-2 | Ochratoxin A (OTA) accumulation assay | Increase in OTA accumulation, impairing OTA efflux through competitive inhibition of MRP-2 pump | Sergent et al. (2005) | |
MCF-7 MX100 | MTX accumulation, topotecan accumulation studies, cytotoxicity assay | Increasing accumulation and inhibiting BCRP in combination with other flavonoids e.g. Biochanin A, increase in accumulation of topotecan (inhibition of the BCRP-mediated transport of topotecan) | Zhang et al. (2004; 2005) | ||
K562/BCRP | Cytotoxicity assay | Strong reversing activity of BCRP-mediated drug resistances | Imai et al. (2004) | ||
L5178/MDR1 | Cytotoxicity assay, Rh-123 accumulation assay | P-gp-mediated efflux pump modifiers (increase in Rh-123 accumulation), strong antiproliferative effects, synergistic effects in combination with epirubicin | Gyémánt et al. (2005) | ||
Chrysosplenol D | Staphylococcus aureus | S. aureus growth inhibition assay | Synergism with berberine and norfloxacin (Potentiated the activity of berberine and norfloxacin against a resistant strain of S. aureus), MDR pump inhibitor |
Stermitz et al. (2002) | |
Diosmetin | K562/BCRP | Cytotoxicity assay | Strong reversing activity of BCRP-mediated drug resistances | Imai et al. (2004) | |
Diosmin | Caco-2 | Accumulation of Rh-123 | Increase in accumulation of Rh-123 | Yoo et al. (2007a) | |
(+)Epicatechin | NIH-3T3-G185 | Intracellular retention of Rh-123 or LDS-751 (P-gp marker substrate) | Slightly facilitated active transport (efflux) of LDS-751 (providing a chemoprotective role) | Wang et al. (2002) | |
(-)Epicatechin | NIH-3T3-G185 | Intracellular retention of Rh-123 or LDS-751 (P-gp marker substrate) | Significant enhance of the active transport (efflux) of LDS-751 (providing a chemoprotective role) | Wang et al. (2002) | |
KB-C2 | Rh-123 and DNR accumulation | No effect on P-gp efflux | Kitagawa et al. (2004) | ||
(-)Epicatechin gallate | NIH-3T3-G185 | Intracellular retention of Rh-123 or LDS-751 (P-gp marker substrate) | Slight inhibition of LDS efflux | Wang et al. (2002) | |
Epicatechin gallate | KB-C2 | Rh-123 and DNR accumulation | Increase in cellular accumulation of Rh-123 and DNR | Kitagawa et al. (2004) | |
Bel-7404/DOX, mouse models | Cell proliferation, Rh-123 and DOX (DOX) accumulation assay, semi-quantitative RT-PCR analysis of MDR1 mRNA expression |
At higher doses a slight inhibitory effect on cell proliferation, administration of DOX with ECG at lower doses significant inhibition of cell proliferation in vitro and hepatoma growth in a xenograft mouse model, increase in DOX and Rh-123 accumulations, decreasing P-gp in cells concurrently treated by DOX and ECG, reduction of the expression of MDR1 mRNA in BEL-7404/DOX cells treated by DOX and ECG | Liang et al. (2010) | ||
(-)Epigallocatechin | NIH-3T3-G185 | Intracellular retention of Rh-123 or LDS-751 (P-gp marker substrate) | Inhibition of LDS efflux | Wang et al. (2002) | |
Epigallocatechin | KB-C2 | Rh-123 and DNR accumulation | Increasing the accumulation of DNR | Kitagawa et al. (2004) | |
MDA-MB-231 | BCECF-AM accumulation assay | MRP-mediated efflux pump modifiers | Gyémánt et al. (2005) | ||
Epigallocatechin gallate | NIH-3T3-G185 | Intracellular retention of Rh-123 or LDS-751 (P-gp marker substrate) | Slight inhibition of the efflux of Rh-123, enhancing the efflux of LDS | Wang et al. (2002) | |
KB-C2 | Rh-123 and DNR accumulation | Increase in Rh-123 and DNR accumulation, decrease in the efflux of Rh-123 | Kitagawa et al. (2004) | ||
Bel-7404/DOX, mouse models | Cell proliferation, Rh-123 and DOX (DOX) accumulation assay, Semi-quantitative RT-PCR analysis of MDR1 mRNA expression |
Slight inhibitory effect on cell proliferation at higher doses, significant inhibition of cell proliferation and hepatoma growth by the administration of DOX with EGCG at lower doses in vitro in a xenograft mouse model, increasing DOX and Rh-123 accumulations, decrease of P-gp in cells concurrently treated by DOX and EGCG, expression of MDR1 mRNA in BEL-7404/DOX cells treated by DOX and EGCG reduced | Liang et al. (2010) | ||
CEM/ADR5000 | MTT assay, Rh-123 accumulation assay using fluorospectroscopy | Reversal of DOX resistance in the cancer cell line | Eid et al. (2013) | ||
Caco-2, CEM/ADR5000 | MTT assay, Rh-123 accumulation assay using fluorospectroscopy | Sensitization of cell lines to DOX | Eid et al. (2013) | ||
MCF-7 | Cytotoxicity using MTS assay, P-gp protein expression using western blot and immunofluorescence microscopy, Rh-123 accumulation | Increase in intracellular Rh-123 accumulation, decrease in P-gp protein expression, reduction of cell viability | Reyes-Esparza et al. (2015) | ||
Caco-2, CEM/ADR5000 | Cytotoxicity determination using MTT assay after combining with DOX, Rh-123 assay and Calcein-AM assay testing P-gp activity | DOX sensitization, synergistic effect on the leukemia cell line | Li et al. (2018b) | ||
Caco-2, CEM/ADR5000 | Cytotoxicity determination using MTT assay after combining with DOX + DTN, Rh-123 assay and Calcein-AM assay testing P-gp activity | DOX sensitization, synergism in both cell lines | Li et al. (2018b) | ||
Formononetin | L5178/MDR1 | Cytotoxicity assay, Rh-123 accumulation assay | P-gp-mediated efflux pump modifier (strong MDR-reversal effects) | Gyémánt et al. (2005) | |
MDA-MB-231 | Cytotoxicity assay, BCECF-AM accumulation assay | MRP-mediated efflux pump modifier, synergistic effects in combination with epirubicin | Gyémánt et al. (2005) | ||
Genistein | K562/BCRP | Topotecan and [3H]Genistein accumulation, cytotoxicity assay | Increasing cytotoxicity of SN-38 and MTX, increasing accumulation of [3H]Genistein and topotecan, inhibition of BCRP-mediated drug efflux | Imai et al. (2004) | |
Panc-1 | Determination of daunomycin and VBL accumulation | Increasing the accumulation of DNM and VBL in Panc-1 cells, inhibiting MRP1-mediated drug transport | Nguyen et al. (2003) | ||
CaCo2 | Ochratoxin A (OTA) accumulation assay | Increase in OTA accumulation, impairing OTA efflux through competitive inhibition of MRP-2 pump | Sergent et al. (2005) | ||
Glabridin | MDCKII | Transport of [3H]digoxin | Inhibition of P-gp-mediated transport of digoxin | Cao et al. (2007) | |
KB-C2 | DNR accumulation | Increase of DNR accumulation (inhibition of the P-gp-mediated efflux of DNR) | Nabekura et al. (2008a) | ||
3,3',4',5,6,7,8-Heptamethoxyflavone | K562/ADM | Uptake of [3H]vincristine | Increasing the uptake of [3H]vincristine (inhibition of P-gp mediated efflux of [3H]vincristine) | Ikegawa et al. (2000) | |
Kaempferol | Panc-1 | Determination of daunomycin and VBL accumulation | Increase in accumulation of DNM and VBL in Panc-1 cells, inhibiting MRP1-mediated drug transport | Nguyen et al. (2003) | |
KB-C2 | Rh-123 and DNR accumulation | Increase in the accumulation of Rh-123 and DNR (inhibition of substrate efflux) | Kitagawa et al. (2005) | ||
K562/BCRP | Cytotoxicity assay | Increasing cytotoxicity of SN-38 and MTX, strong reversing activity of BCRP-mediated drug resistances | Imai et al. (2004) | ||
MDA-MB-231 | Cytotoxicity assay, BCECF-AM accumulation assay | MRP-mediated efflux pump modifier, synergistic effects in combination with epirubicin | Gyémánt et al. (2005) | ||
Luteolin | K562/BCRP | Cytotoxicity assay | Strong reversing activity of BCRP-mediated drug resistances | Imai et al. (2004) | |
Morin | MDA435/LCC6, MCF-7/ADR, MDA435/LCCMDR1 | Daunomycin (DNM) accumulation, P-gp ATPase activity assay, [3H]azidopine photoaffinity labeling | Increase in DNM accumulation, inhibition of P-gp ATPase activity, inhibition of P-gp-mediated cellular efflux, inhibition of [3H]azidopine photoaffinity labeling of P-gp suggesting a direct interaction with P-gp substrate binding | Zhang and Morris (2003) | |
Panc-1 | Determination of daunomycin and VBL accumulation | Increasing the accumulation of DNM and VBL in Panc-1 cells, inhibiting MRP1-mediated drug transport | Nguyen et al. (2003) | ||
Myricetin | MDCKII-MRP1, MDCKII-MRP2 | Efflux of calcein (inhibition of MRP1 and MRP2 activity was studied using the fluorescent calcein as a model substrate |
Inhibition of MRP1 and MRP2 activity (inhibition of calcein efflux) | van Zanden et al. (2005) | |
Naringenin | MCF-7/ADR | [3H]-Daunomycin (DNM) accumulation, [3H]-Daunomycin efflux study | Increase in accumulation of DNM, decrease in efflux of DNM | Chung et al. (2005) | |
Caco-2 | The flux of talinolol across Caco-2 cell monolayers | Reduction of P-gp mediated secretory transport of talinolol (inhibition of P-gp) | De Castro et al. (2008) | ||
K562/BCRP | Cytotoxicity assay, topotecan accumulation | Increase in cytotoxicity of SN-38 and MTX, increase in accumulation of topotecan | Imai et al. (2004) | ||
Naringenin-7-glucosid | K562/BCRP | Cytotoxicity assay | Inhibition of BCRP-mediated drug resistance | Imai et al. (2004) | |
Nobiletin | KB-C2, KB/MRP | Accumulation assay of DNR in KB-C2 cells and calcein in KB/MRP cells, P-gp ATPase activity | Increase in the accumulation of DNR in KB-C2 cells, increase in calcein accumulation in KB/MRP cells, stimulation of ATPase activity of P-gp | Nabekura et al. (2008b) | |
K562/ADM | Uptake of [3H]vincristine | Increase in the uptake of [3H]vincristine (inhibition of P-gp mediated efflux of [3H]vincristine) | Ikegawa et al. (2000) | ||
A2780/T, A549/T | Cytotoxicity assay (SRB assay); intracellular accumulation of Rh-123, DOX and Flutax-2 using flow cytometry | Sensitization of cells to chemotherapeutic drugs PTX, DOX, DNR and docetaxel; synergism with PTX; increase in intracellular accumulation of Rh-123, DOX and Flutax-2 | Ma et al. (2015) | ||
Phloretin | MDA435/LCC6, MCF-7/ADR | Daunomycin (DNM) accumulation | Increase in DNM accumulation, inhibition of P-gp-mediated cellular efflux | Zhang and Morris (2003) | |
Panc-1 | Determination of daunomycin and VBL accumulation | Increase in the accumulation of DNM and VBL in Panc-1 cells, inhibiting MRP1-mediated drug transport | Nguyen et al. (2003) | ||
Mouse lymphoma/MDR1 cells | Rh-123 accumulation | Moderate inhibition of efflux | Molnár et al., 2010 (review) | ||
Procyanidine | Rat brain microvessel endothelial cells (RBMECs) | Rh-123 intracellular accumulation assay, Rh-123 efflux assay, P-gp ATPase activity measurement | Increase in the accumulation of Rh-123, decrease in Rh-123 efflux, inhibition of the P-gp ATPase activity | He et al. (2009) | |
Quercetin | Panc-1 | Determination of daunomycin and VBL accumulation | Increase in the accumulation of DNM and VBL in Panc-1 cells, inhibition of MRP1-mediated drug transport | Nguyen et al. (2003) | |
CaCo2 | Ochratoxin A (OTA) accumulation assay | Increase of OTA accumulation, impairing OTA efflux through competitive inhibition of MRP-2 pump | Sergent et al. (2005) | ||
BEL/5-FU | Cytotoxicity assay; Rh-123 and ADR accumulation using flow cytometry; ABCB1, ABCC1, ABCC2 mRNA and protein expression using real-time PCR and western blot | Increase in sensitivity to chemotherapeutic drugs 5-FU, MMC and ADR; increase in intracellular Rh-123 and ADR accumulation; decrease in ABCB1, ABCC1 and ABCC2 mRNAs and proteins expression | Chen et al. (2018) | ||
Robinetin | MDCKII-MRP1, MDCKII-MRP2 | Efflux of calcein (inhibition of MRP1 and MRP2 activity was studied using the fluorescent calcein as a model substrate |
Inhibition of MRP1 and MRP2 activity (inhibited calcein efflux) | van Zanden et al. (2005) | |
Robinin | MDA-MB-231 | Cytotoxicity assay, BCECF-AM accumulation assay | MRP-mediated efflux pump modifiers, additive effects in combination with epirubicin | Gyémánt et al. (2005) | |
Rotenone | Mouse lymphoma/MDR1, COLO320/MDR1 |
Rh-123 accumulation | Inhibited efflux, increased Rh-123 accumulation | Molnár et al., 2010 (review) | |
L5178/MDR1 | Cytotoxicity assay, Rh-123 accumulation assay | P-gp-mediated efflux pump modifier (MDR modulating activity), strong antiproliferative effects, additive effects in combination with epirubicin | Gyémánt et al. (2005) | ||
Silymarin | MCF-7/ADR | [3H]-Daunomycin (DNM) accumulation, [3H]-Daunomycin efflux study | Increase in the accumulation of DNM, decrease in efflux of DNM | Chung et al. (2005) | |
MDA435/LCC6, MCF-7/ADR, MDA435/LCCMDR1 | Daunomycin (DNM) accumulation, P-gp ATPase activity assay, [3H]azidopine photoaffinity labeling | Increase in DNM accumulation, inhibition of P-gp ATPase activity, inhibition of P-gp-mediated cellular efflux, inhibition of [3H]azidopine photoaffinity labeling of P-gp suggesting a direct interaction with the P-gp substrate binding | Zhang and Morris (2003) | ||
Panc-1 | Determination of daunomycin and VBL accumulation | Increase in the accumulation of DNM and VBL in Panc-1 cells, inhibition of MRP1-mediated drug transport | Nguyen et al. (2003) | ||
Tangeretin | K562/ADM | Uptake of [3H]vincristine | Increase in the uptake of [3H]vincristine (inhibition of P-gp mediated efflux of [3H]vincristine) | Ikegawa et al. (2000) | |
Stilbenoids | Resveratrol | KB-C2 | Determination of DNR and Rh-123 accumulation | Increase in the accumulation of DNR, decrease in the efflux of Rh-123 | Nabekura et al. (2005) |
CaCo2 | Ochratoxin A (OTA) accumulation assay | Increase of OTA accumulation, impairing OTA efflux through competitive inhibition of MRP-2 pump | Sergent et al. (2005) | ||
Caco-2, CEM/ADR5000 | Cytotoxicity assay using MTT, Rh-123 accumulation assay | Increase in Rh-123 accumulation, enhancement of DOX cytotoxicity | El-Readi et al. (2019) | ||
Curcuminoids | Curcumin | SGC7901/VCR | Analysis of apoptosis by propidium iodide (PI)-stained flow cytometry (FCM) and a morphological assay using acridine orange (AO)/ethidium bromide (EB) dual staining, accumulation and efflux of Rh123 as measured by flow cytometry, expression of P-gp by FCM using fluorescein isothiocyanate (FITC)-conjugated anti-P-gp |
Promotion of VCR-mediated apoptosis, increase in Rh-123 accumulation and inhibition of the efflux of Rh-123, downregulation of P-gp expression | Tang et al. (2005) |
KBV20C | VCR, PTX | Increasing cytotoxicity | Um et al. (2008) | ||
KB-C2 | Determination of DNR and Rh-123 accumulation | Increase in the accumulation of DNR and Rh-123 | Nabekura et al. (2005) | ||
Caco-2, LLC-PK1, LLC-GA5-COL300 | DNR transport (apical to basolateral (a-b) and basolateral to apical (b-a)) across Caco-2 cell monolayers, calcein-AM uptake in LLC-PK1 and LLC-GA5-COL300 cells | Decrease in the efflux ratio of DNR, increase of calcein-AM accumulation in LLC-GA5-COL300 | Ampasavate et al. (2010) | ||
Caco-2, CEM/ADR5000 | Cytotoxicity determination using MTT assay after combining with DOX, Rh-123 assay and Calcein-AM assay testing P-gp activity | DOX sensitization, synergistic effect on the colon cancer cell line | Li et al. (2018b) | ||
Caco-2, CEM/ADR5000 | Cytotoxicity determination using MTT assay after combining with DOX + DTN, Rh-123 assay and Calcein-AM assay testing P-gp activity | DOX sensitization, synergism in both cell lines | Li et al. (2018b) | ||
Bisdemethoxycurcumin | Caco-2, LLC-PK1, LLC-GA5-COL300 | DNR transport (apical to basolateral (a-b) and basolateral to apical (b-a)) across Caco-2 cell monolayers, calcein-AM uptake in LLC-PK1 and LLC-GA5-COL300 cells | Decrease in the efflux ratio of DNR | Ampasavate et al. (2010) | |
Demethoxycurcumin, | Caco-2, LLC-PK1, LLC-GA5-COL300 | DNR transport (apical to basolateral (a-b) and basolateral to apical (b-a)) across Caco-2 cell monolayers, calcein-AM uptake in LLC-PK1 and LLC-GA5-COL300 cells | Decrease in the efflux ratio of DNR, increase of calcein-AM accumulation in LLC-GA5-COL300 | Ampasavate et al. (2010) | |
Tetrahydrocurcumin | KB-V1, MCF-7 MDR | Rh-123 and calcein-AM accumulation assay by FACS in KB-V1 cells, radiolabeled drug ([3H]-VBL) accumulation for MCF-7 MDR | Increase in the accumulation of Rh-123 and calcein-AM in KB-V-1 cells, increase in the accumulation and inhibition of the [3H]-VBL efflux in MCF-7 MDR | Limtrakul et al. (2007) | |
Others | Tannic acid | Caco-2, CEM/ADR5000 | Cytotoxicity determination using MTT assay after combining with DOX, Rh-123 assay and Calcein-AM assay testing P-gp activity | DOX sensitization, synergistic effect on the colon cancer cell line | Li et al. (2018b) |
Caco-2, CEM/ADR5000 | Cytotoxicity determination using MTT assay after combining with DOX + DTN, Rh-123 assay and Calcein-AM assay testing P-gp activity | DOX sensitization, synergism in both cell lines | Li et al. (2018b) | ||
Phenylpropanoids | |||||
Neo-/lignans | Arctigenin | Caco-2 | Cytotoxicity determination using MTT assay after combining with DOX, Rh-123 accumulation assay testing P-gp activity | Synergism in Caco-2 cells and slight synergistic effect in CEM/ADR5000, increase in Rh-123 accumulation | Su et al. (2015) |
Caco-2, CEM/ADR5000 | Cytotoxicity determination using MTT assay after combining with DOX + DTN, Rh-123 accumulation assay testing P-gp activity | Significant increase in synergism of combination with DOX by DTN, increase in Rh-123 accumulation | Su et al. (2015) | ||
Arctiin | Caco-2, CEM/ADR 5000 | Cytotoxicity determination using MTT assay after combining with DOX, Rh-123 accumulation assay testing P-gp activity | Moderate synergistic effect in CEM/ADR (concentration-dependent) and in Caco-2 cells, increase in Rh-123 accumulation | Su et al. (2015) | |
Cytotoxicity determination using MTT assay after combining with DOX + DTN, Rh-123 accumulation assay testing P-gp activity | Significant increase in synergism of combination with DOX by DTN, increase in Rh-123 accumulation | Su et al. (2015) | |||
Epimagnolin A | Flp-In-293/ABCB1 | MTT assay; calcein assay | Enhancement of sensitivity to anti-cancer drugs DNR, DOX, VBL and VCR; inhibition of calcein efflux | Mitani et al. (2018) | |
Iso-/lappaol A | Caco-2 | Cytotoxicity determination using MTT assay after combining with DOX, Rh-123 accumulation assay testing P-gp activity | Slight synergistic effect in Caco-2 cells, additive effect in CEM/ADR cells, increase in Rh-123 accumulation | Su et al. (2015) | |
Caco-2, CEM/ADR5000 | Cytotoxicity determination using MTT assay after combining with DOX + DTN, Rh-123 accumulation assay testing P-gp activity | Significant increase in synergism of combination with DOX by DTN, increase in Rh-123 accumulation | Su et al. (2015) | ||
Lappaol C | Caco-2, CEM/ADR 5000 | Cytotoxicity determination using MTT assay after combining with DOX, Rh-123 accumulation assay testing P-gp activity | Synergistic effect in CEM/ADR (concentration-dependent) and in Caco-2 cells | Su et al. (2015) | |
Caco-2, CEM/ADR 5000 | Cytotoxicity determination using MTT assay after combining with DOX + DTN, Rh-123 accumulation assay testing P-gp activity | Significant increase in synergism of combination with DOX by DTN | Su et al. (2015) | ||
Lappaol F | Caco-2, CEM/ADR 5000 | Cytotoxicity determination using MTT assay after combining with DOX, Rh-123 accumulation assay testing P-gp activity | Moderate synergism in CEM/ADR 5000 cells with concentration-dependent activity, stronger effect in Caco-2 cells, increase in Rh-123 accumulation | Su et al. (2015) | |
Cytotoxicity determination using MTT assay after combining with DOX + DTN, Rh-123 accumulation assay testing P-gp activity | Significant increase in synergism of combination with DOX by DTN, increase in Rh-123 accumulation | Su et al. (2015) | |||
Matairesinol | KB-C2, KB/MRP | DNR and calcein accumulation assay | Increasing accumulation of DNR (inhibits the P-gp-mediated efflux of DNR) and calcein (inhibits the MRP1-mediated efflux of calcein) | Nabekura et al. (2008a) | |
Caco-2, CEM/ADR 5000 | Cytotoxicity determination using MTT assay after combining with DOX, Rh-123 accumulation assay testing P-gp activity | Synergistic effect in Caco-2 cells, increase in Rh-123 accumulation, reversal of multidrug resistance | Su et al. (2015) | ||
Caco-2, CEM/ADR 5000 | Cytotoxicity determination using MTT assay after combining with DOX + DTN, Rh-123 accumulation assay testing P-gp activity | Significant increase in synergism of combination with DOX by DTN, increase in Rh-123 accumulation | |||
Sesamin | KB-C2 | DNR accumulation assay | Increase in DNR accumulation (inhibition of the P-gp-mediated efflux of DNR) | Nabekura et al. (2008a) | |
Dibenzocyclo-octadienelignans | Gomisin A | HepG2-DR | Cellular Rh-123 accumulation assay by flow cytometry, determination of P-gp-associated ATPase activity, photoaffinity labeling of P-gp with [125I]iodoarylazidoprazosin | Restoration of the cytotoxicity of VBL and DOX, inhibition of the P-gp ATPase activity, additive effect with verapamil and vanadate on the inhibition of Rh-123 efflux, inhibition of [125I]IAAP photo-crosslinking of P-gp | Wan et al. (2006) |
Schisandrin A (Deoxyschizandrin) | Caco-2 | Rh-123 uptake assay, bidirectional transports of digoxin and Rh-123 | Increase in Rh-123 accumulation, increase in apical-to-basal transports of digoxin and Rh-123, decrease in basal-to-apical transports | Yoo et al. (2007b) | |
COR-L23/R | MTT assay, DOX accumulation assay | Restoration of the cytotoxic action of DOX to COR-L23/R cells, increase in the accumulation of DOX | Slaninová et al. (2009) | ||
Schisandrin B/γ-Schizandrin | COR-L23/R | MTT assay, DOX accumulation assay | Restoration of the cytotoxic action of DOX to COR-L23/R cells, increase in the accumulation of DOX | Slaninová et al. (2009) | |
Schisandrol A | HepG2-DR | Flow cytometry analyses of cell cycle and Rh-123 efflux, P-gp-ATPase activity assay | Strong synergistic effect (enhanced cytotoxicity) with DOX, VBL and taxol, restoration VBL-induced G2/M arrest, increase in cellular retention of Rh-123, stimulation of basal P-gp-ATPase | Fong et al. (2007) | |
Others | Chlorogenic acid | Rats jejunal membrane in vitro | P-gp stimulation/inhibition profiles using a P-gp-dependent ATPase assay | Inhibitory effect on P-gp ATPase activity | Najar et al. (2010) |
Ginkgolic acid | KB-C2 | DNR accumulation | Increase of DNR accumulation (inhibiting the P-gp-mediated efflux of DNR) | Nabekura et al. (2008a) | |
Terpenes | |||||
Monoterpenes | Citronellal | LLC-GA5-COL150 | Intracellular accumulation of [3H]digoxin | Increase in [3H]digoxin accumulation | Yoshida et al. (2005) |
(R)-(+)-citronellal | LLC-GA5-COL150 | Intracellular accumulation of [3H]digoxin | Increase in [3H]digoxin accumulation | Yoshida et al. (2005) | |
(S)-(–) β-citronellol | LLC-GA5-COL150 | Intracellular accumulation of [3H]digoxin | Increase in [3H]digoxin accumulation | Yoshida et al. (2005) | |
Cineole | LLC-GA5-COL150 | Intracellular accumulation of [3H]digoxin | Increase in [3H]digoxin accumulation | Yoshida et al. (2005) | |
DL-citronellol | LLC-GA5-COL150 | Intracellular accumulation of [3H]digoxin | Increase in [3H]digoxin accumulation | Yoshida et al. (2005) | |
Menthol | Caco-2, CEM/ADR5000 | MTT assay, Rh-123 accumulation assay using fluorospectroscopy | Reversal of DOX resistance in both cell lines, synergism with DOX | Eid et al. (2013) | |
Caco-2, CEM/ADR5000 | MTT assay, Rh-123 accumulation assay using fluorospectroscopy | Synergism with DOX | Eid et al. (2013) | ||
Thymol | Caco-2, CEM/ADR5000 | MTT assay, Rh-123 accumulation assay using fluorospectroscopy | Synergism with DOX | Eid et al. (2013) | |
Caco-2, CEM/ADR5000 | MTT assay, Rh-123 accumulation assay using fluorospectroscopy | Synergism with DOX | Eid et al. (2013) | ||
Iridoid glucosides | Agnuside | Rats jejunal membrane in vitro | P-gp stimulation/inhibition profiles using a P-gp-dependent ATPase assay | Inhibitory effect on P-gp ATPase activity | Najar et al. (2010) |
Negundoside | Rats jejunal membrane in vitro | P-gp stimulation/inhibition profiles using a P-gp-dependent ATPase assay | Stimulatory effect on P-gp ATPase activity | Najar et al. (2010) | |
Picroside-I | Rats jejunal membrane in vitro | P-gp stimulation/inhibition profiles using a P-gp-dependent ATPase assay | Stimulatory effect on P-gp ATPase activity | Najar et al. (2010) | |
Picroside-II | Rats jejunal membrane in vitro | P-gp stimulation/inhibition profiles using a P-gp-dependent ATPase assay | Inhibitory effect on P-gp ATPase activity | Najar et al. (2010) | |
Sesquiterpenes | Aromadendrene | Caco-2, CEM/ADR5000 | MTT assay using, Rh-123 accumulation assay using fluorospectroscopy | Synergism with DOX | Eid et al. (2013) |
Caco-2, CEM/ADR5000 | MTT assay using, Rh-123 accumulation assay using fluorospectroscopy | Synergism with DOX | Eid et al. (2013) | ||
Farnesiferol B | MCF-7/Adr | Cytotoxicity using alamar blue assay, accumulation of Rh-123 using flow cytometry | Increase in cytotoxicity of DOX, increase in intracellular accumulation of Rh-123 | Kasaian et al. (2015) | |
Farnesiferol C | MCF-7/Adr | Cytotoxicity using alamar blue assay, accumulation of Rh-123 using flow cytometry | Increase in cytotoxicity of DOX, increase in intracellular accumulation of Rh-123 | Kasaian et al. (2015) | |
Lehmferin | MCF-7/Adr | Cytotoxicity using alamar blue assay, accumulation of Rh-123 using flow cytometry | Increase in cytotoxicity of DOX, increase in intracellular accumulation of Rh-123 | Kasaian et al. (2015) | |
Santonin | Rats jejunal membrane in vitro | P-gp stimulation/inhibition profiles using a P-gp-dependent ATPase assay | Inhibitory effect on P-gp ATPase activity | Najar et al. (2010) | |
Umbelliprenin | MCF-7/Adr | Cytotoxicity using alamar blue assay, accumulation of Rh-123 using flow cytometry | Increase in cytotoxicity of DOX, increase in intracellular accumulation of Rh-123 | Kasaian et al. (2015) | |
Diterpenes | Andrographolide | Rats jejunal membrane in vitro | P-gp stimulation/inhibition profiles using a P-gp-dependent ATPase assay | Biphasic effect (stimulation at low concentration and inhibition at high concentration) | Najar et al. (2010) |
Carnosic acid | KB-C2, KB/MRP | Accumulation assay of DNR and Rh-123 in KB-C2 and calcein in KB/MRP cells, ATPase activity assay, resistance to VBL cytotoxicity by a calorimetric assay | Increase in the accumulation of DNR and Rh-123 in KB-C2 cells, stimulation of the P-gp ATPase activity, sensitization of KB-C2 cells to VBL cytotoxicity | Nabekura et al. (2010) | |
Carnosol | KB-C2, KB/MRP | Accumulation assay of DNR and Rh-123 in KB-C2 and calcein in KB/MRP cells, ATPase activity assay, resistance to VBL cytotoxicity by a calorimetric assay | Increase in the accumulation of DNR and Rh-123 in KB-C2 cells, stimulation of P-gp ATPase activity | Nabekura et al. (2010) | |
Esulatin M | L5178Y‐MDR | MTT assay; Rh-123 accumulation assay | Synergistic interaction with DOX; increase in intracellular accumulation of Rh-123 | Reis et al. (2016) | |
Epoxywelwitschene | L5178Y‐MDR | MTT assay; Rh-123 accumulation assay | Synergistic interaction with DOX; increase in intracellular accumulation of Rh-123 | Reis et al. (2016) | |
Euphoboetirane A, C, D, E, F, G and I | L5178Y‐MDR | MTT assay; Rh‐123 accumulation assay | Increase in intracellular Rh-123 accumulation; synergism with DOX | Neto et al. (2019) | |
Euphotuckeyanol | L5178/MDR1 | Rh-123 accumulation assay, in vitro antiproliferative effect in combination with epirubicin using checkerboard microplate method | Inhibition of Rh-123 efflux (increase in accumulation), exhibition of a synergistic interaction with epirubicin and enhancement of the antiproliferative effect of epirubicin | Duarte et al. (2008) | |
Euphowelwitschine A, B | L5178Y‐MDR | MTT assay; Rh-123 accumulation assay | Synergistic interaction with DOX; increase in intracellular accumulation of Rh-123 | Reis et al. (2016) | |
12‐Hydroxyboetirane A, B and C | L5178Y‐MDR | MTT assay; Rh‐123 accumulation assay | Increase in intracellular Rh-123 accumulation; synergism with DOX | Neto et al. (2019) | |
Latilagascene A, B, C | L5178/MDR1 | Assay for Rh-123 accumulation | Inhibition of Rh-123 efflux (increase in accumulation) | Duarte et al. (2006) | |
Latilagascene G, H, I | L5178/MDR1 | Rh-123 accumulation assay, in vitro antiproliferative effect in combination with epirubicin using checkerboard microplate method | Inhibition of Rh-123 efflux (increase in accumulation), exhibition of a synergistic interaction by all compounds with epirubicin and enhancement of the antiproliferative effect of epirubicin | Duarte et al. (2008) | |
Totarol | Staphylococcus aureus SA-K3092 (Totarol-resistant mutant overexpressing norA) | Checkerboard combination studies using ethidium bromide (EtBr) and totarol, EtBr efflux assay, modulatory activity of totarol at half the MIC | Reduction of the MICs of selected antibiotics by subinhibitory concentrations (suggesting that it may be an efflux pump inhibitor), reduction of ethidium efflux and ethidium MIC in SA-K3092 | Smith et al. (2007) | |
Tuckeyanol A, B | L5178/MDR1 | Rh-123 accumulation assay, in vitro antiproliferative effect in combination with epirubicin using checkerboard microplate method | Inhibition of Rh-123 efflux (increase in accumulation), exhibition of a synergistic interaction by both compounds with epirubicin and enhancement of the antiproliferative effect of epirubicin | Duarte et al. (2008) | |
Welwitschene | L5178Y‐MDR | MTT assay; Rh-123 accumulation assay | Increase in intracellular accumulation of Rh-123 | Reis et al. (2016) | |
Triterpenoids, Saponins* | β-Amyrin | MDR1-transfected mouse lymphoma cells | Accumulation of Rh-123, accumulation of ethidium bromide (semi-automated ethidium bromide fluorometric method) | Increase in the accumulation of ethidium bromide (inhibitory activity against the P-gp transporter) | Martins et al. (2010) |
Cumingianol A, B, D | KB-C2 | Cytotoxicity assay (MTT) | Enhancement of cytotoxicity against KB-C2 cells in the presence of colchicine | Kurimoto et al. (2011a) | |
Cumingianol D | MCF7 | Cytotoxicity assay (MTT) | Moderate cytotoxicity | Kurimoto et al. (2011a) | |
Deacetylnomilin | CEM/ADR5000, Caco-2 | Measurement of DOX cytotoxicity (reversal assay), Rh-123 efflux assay | Inhibition of Rh-123 efflux in CEM/ADR5000 cells, increase in DOX cytotoxicity in Caco-2 cells | El-Readi et al. (2010) | |
Dyscusin A | KB-C2 | Cytotoxicity assay (MTT) | Enhancement of cytotoxicity against KB-C2 cells in the presence of colchicine | Kurimoto et al. (2011b) | |
Glycyrrhetinic acid (Enoxolone) | KB-C2, KB/MRP | DNR and calcein accumulation | Increasing DNR (inhibits the P-gp-mediated efflux of DNR) and calcein (inhibits the MRP1-mediated efflux of calcein) accumulation | Nabekura et al. (2008a) | |
Glycyrrhizin* | Rats jejunal membrane in vitro | P-gp stimulation/inhibition profiles using a P-gp-dependent ATPase assay | Biphasic effect (stimulation at low concentration and inhibition at high concentration) | Najar et al. (2010) | |
Limonin | CEM/ADR5000, Caco-2 | Measurement of DOX cytotoxicity (reversal assay), Rh-123 efflux assay | Inhibition of Rh-123 efflux in CEM/ADR5000 cells, enhancement of DOX cytotoxicity in CEM/ADR5000 and Caco-2 cells | El-Readi et al. (2010) | |
Obacunone | MES-SA/DX5, HCT15 | Cytotoxicity assay in the presence of PTX | Significant inhibition of the P-gp MDR activity | Min et al. (2007) | |
Oleanolic acid | Rats jejunal membrane in vitro | P-gp stimulation/inhibition profiles using a P-gp-dependent ATPase assay | Stimulatory effect on P-gp ATPase activity | Najar et al. (2010) | |
MDR1-transfected mouse lymphoma cells | Accumulation of Rh-123, accumulation of ethidium bromide (semi-automated ethidium bromide fluorometric method) | Increase in the accumulation of ethidium bromide (inhibitory activity against the P-gp transporter) | Martins et al. (2010) | ||
Sinocalycanchinensin E | KB-C2 | Cytotoxicity assay (MTT) | Enhancement of the cytotoxicity against KB-C2 cells in the presence of colchicine | Kashiwada et al. (2011) | |
Ursolic Acid | KB-C2, KB/MRP | Accumulation assay of DNR and Rh-123 in KB-C2 and calcein in KB/MRP cells, ATPase activity assay, resistance to VBL cytotoxicity by a calorimetric assay | Increase in the accumulation of DNR and Rh-123 in KB-C2 cells, stimulation of P-gp ATPase activity | Nabekura et al. (2010) | |
Uvaol | MDR1-transfected mouse lymphoma cells | Accumulation of Rh-123, accumulation of ethidium bromide (semi-automated ethidium bromide fluorometric method), checker-board assay (for the study of synergism between uvaol and DOX) | Increase in the accumulation of Rh-123 and ethidium bromide (inhibitory activity against the P-gp transporter), synergism with DOX cytotoxicity | Martins et al. (2010) | |
Steroids, Saponins* | Alisol B 23-acetate | HepG2-DR, K562-DR | Cellular Rh-123 and DOX accumulation, photoaffinity labeling of P-gp with [125I]iodoarylazidoprazosin, P-gp ATPase activity, | Enhancement of VBL toxicity, restoration of the activity of VBL in causing G2/M arrest, increase in DOX accumulation, delay of Rh-123 efflux, inhibition of the photoaffinity labeling of P-gp by [125I]IAAP, stimulation of the P-gp ATPase activity | Wang et al. (2004) |
11α-O-benzoyl-12β-O-acetyltenacigenin B | HepG2/DOX | MDR reversing potential evaluation (comparing IC50 values of an anticancer drug in the absence or presence of 11-Alpha-O-benzoyl-12β-O-acetyl-tenacigenin B) | Increase in the sensitivity of HepG2/Dox cells to the antitumor drugs DOX, VBL, puromycin, and PTX | Hu et al. (2008) | |
DTN | Caco-2, CEM/ADR5000 | MTT assay using, Rh-123 accumulation assay using fluorospectroscopy | Reversal of DOX resistance, reduction of the IC50 value of DOX | Eid et al. (2013) | |
Ginsenoside M1, M4 and M12 (hydrolyzed metabolites of ginsenoside) | KB-C2 | DNR accumulation, verapamil-induced ATPase activity (for M4) | Increase in DNR accumulation by M1, M4 and M12; decrease of ATPase activity by M4 (most potent substance in this study) | Kitagawa et al. (2007) | |
Ginsenoside Rc and Rd | Multidrug resistant mouse lymphoma cells | – | Moderate reduction of the activity of the efflux pump | Berek et al. (2001) | |
Ginsenoside Rg3* | KBV20C | Rh-123 retention assay, photo-affinity labeling with [3H]azidopine, [3H]VBL accumulation, cytotoxicity assay using Sulforhodamine B cell staining method | Increase in accumulation of Rh-123, inhibition of [3H]VBL efflux, prevention of binding of [3H]azidopine to P-gp, restoration of the sensitivity of KBV20C cells to DOX, COL, VCR, and VP-16 | Kim et al. (2003) | |
Ginsenosides Rg1, Rc, Rd, Re* | Multidrug resistant mouse lymphoma cells | – | Moderate inhibitory effect on the drug efflux pump | Molnár et al. (2000) | |
Guggulsterone | KB-C2, KB/MRP | Accumulation assay of DNR and Rh-123 in KB-C2 cells and calcein in KB/MRP cells, ATPase activity of P-gp and MRP1 | Increase in Rh-123 and DNR accumulation in KB-C2 cells, inhibition of Rh-123 efflux from KB-C2 cells, increase in the accumulation of calcein in KB/MRP cells, stimulation of ATPase activities of P-gp and MRP1 | Nabekura et al. (2008c) | |
Methylprototribestin* | L5178/MDR1 | Rh-123 accumulation, checkerboard microplate method to study the interaction (MDR reversal effect) between the methylprototribestin and DOX | Increase in Rh-123 accumulation, synergistic interaction between methylprototribestin and DOX | Ivanova et al. (2009) | |
Protopanaxatriol ginsenosides* | AML-2/D100 | Accumulation assay of DNR, [3H]-azidopine photolabeling of P-gp | Reversing the resistance to DNR, increase in DNR accumulation, inhibition of [3H]-azidopine photolabeling of P-gp | Choi et al. (2003) | |
β-sitosterol-O-glucoside* | CEM/ADR5000, Caco-2 | Measurement of DOX cytotoxicity (reversal assay), Rh-123 efflux assay | Inhibition of Rh-123 efflux in CEM/ADR5000 cells, increase in the cytotoxicity of DOX in Caco-2 cells | El-Readi et al. (2010) | |
Caco-2, CEM/ADR5000 | MTT assay, Rh-123 accumulation assay using fluorospectroscopy | Reversal of DOX resistance in both cell lines, synergism | Eid et al. (2013) | ||
Caco-2, CEM/ADR5000 | MTT assay, Rh-123 accumulation assay using fluorospectroscopy | Sensitization of cell lines, enhancement of cytotoxicity, light reduction of the IC50 value of DOX, synergism in the Caco-2 cells | Eid et al. (2013) | ||
Stigmasterol | Caco-2, CEM/ADR5000 | Measurement of DOX cytotoxicity (reversal assay), Rh-123 efflux assay | Inhibition of Rh-123 efflux in CEM/ADR5000 cells, increase in the cytotoxicity of DOX in Caco-2 cells | El-Readi et al. (2010) | |
Tenacigenin B: P8, P26 and P27 | SW620/Ad300 (P-gp-overexpressing) MCF-7/VP (MRP-1-overexpressing) MCF-7/FLV1000 (ABCG2-overexpressing) |
MDR reversal effect on cytotoxicity of anticancer drugs DOX (substrate of P-gp and MRP1) and MTX (substrate of ABCG2), flow cytometry-based efflux assay of Rh-123 (P-gp substrate), calcein AM (MRP1 substrate) and PhA (ABCG2 substrate) | Effective in circumventing MDR mediated by P-gp, MRP1 and ABCG2, inhibition of P-gp efflux activity, increase in intracellular concentration of the substrate drugs through the inhibition of MRP1- and ABCG2-mediated efflux | To et al. (2017) | |
Tenacigenin B: P2, P3 and P6 | SW620/Ad300 (P-gp-overexpressing), MCF-7/VP (MRP-1-overexpressing), MCF-7/FLV1000 (ABCG2-overexpressing) |
MDR reversal effect on cytotoxicity of anticancer drugs DOX (substrate of P-gp and MRP1) and MTX (substrate of ABCG2), flow cytometry-based efflux assay of Rh-123 (P-gp substrate), calcein AM (MRP1 substrate) and PhA (ABCG2 substrate) | Effective in circumventing MDR mediated by P-gp and MRP1, inhibition of P-gp efflux activity by P2 and P6, increase in the intracellular concentration of the substrate drug via inhibition of MRP1-mediated efflux | To et al. (2017) | |
Tenacigenin B: P1, P4, P5, P9 and P28 | SW620/Ad300 (P-gp-overexpressing) MCF-7/VP (MRP-1-overexpressing) MCF-7/FLV1000 (ABCG2-overexpressing) |
MDR reversal effect on cytotoxicity of anticancer drugs DOX (substrate of P-gp and MRP1) and MTX (substrate of ABCG2), flow cytometry-based efflux assay of Rh-123 (P-gp substrate), calcein AM (MRP1 substrate) and PhA (ABCG2 substrate) | Effective in circumventing P-gp-mediated MDR, P1 and P5 inhibition of P-gp efflux activity | To et al. (2017) | |
Tenacissimoside A* | HepG2/DOX | DOX accumulation, Rh-123 and Hoechst 33342 efflux assay, cell cycle analysis, MDR reversing potential evaluation (comparing IC50 values of an anticancer drug in the absence or presence of Tenacissimoside A) | Increase in the sensitivity of HepG2/Dox cells to the antitumor drugs DOX, VBL, puromycin, and PTX, increase in DOX accumulation, enhancement of the action of DOX in causing G2/M arrest, inhibition of the efflux of Rh-123 and Hoechst 33342 | Hu et al. (2008) | |
Tetraterpenes | Aurochrome | L5178/MDR1 | Rh-123 accumulation assay | Moderate inhibition of P-gp | Gyémánt et al. (2006) |
Canthaxanthin | Caco-2, CEM/ADR5000 | MTT assay, activity determination using Rh-123- and Calcein-AM retention assay | Significant enhancement of cytotoxicity and synergism using the following drugs: PTX, Cycloheximide, DOX, VBL, amphotericin-B, 5-FU, etoposide and cisplatine |
Eid et al. (2012) | |
Capsanthin | MDR1 gene-transfected mouse lymphoma (L1210) cells | Rh-123 accumulation assay | Inhibition of the efflux, very active in MDR reversal effect | Molnár et al. (2006) | |
Capsorubin | Human MDR1-gene-transfected mouse lymphoma (L1210) cells | Rh-123 accumulation assay | MDR reversal activity, enhancement of Rh-123 accumulation | Molnár et al. (2006) | |
β-carotene | Caco-2, CEM/ADR5000 | MTT assay, activity determination using Rh-123- and Calcein-AM retention assay | Synergism, significant enhancement of cytotoxicity of DOX, VBL, amphotericin-B, 5-FU, etoposide and cisplatine; substrate |
Eid et al. (2012) | |
Caco-2, CEM/ADR5000 | MTT assay using, Rh-123 accumulation assay using fluorospectroscopy | Sensitization of cell lines, enhancement of cytotoxicity, strong reduction of the IC50 value of DOX and consequently increase of efficacy, synergism | Eid et al. (2013) | ||
Caco-2, CEM/ADR5000 | MTT assay using, Rh-123 accumulation assay using fluorospectroscopy | Reversal of DOX resistance in both cell lines, synergism | Eid et al. (2013) | ||
ABCB1/Flp-InTM-293 | MTT assay; calcein-AM accumulation assay | Increase in cytotoxicity of DOX; increase in intracellular concentration of calcein | Teng et al. (2016) | ||
Crocin | Caco-2, CEM/ADR5000 | MTT assay, activity determination using Rh-123- and Calcein-AM retention assay | Synergism, enhancement of cytotoxicity of DOX, VBL, cisplatine; substrate | Eid et al. (2012) | |
Diepoxycarotene | L5178/MDR1 | Rh-123 accumulation assay | Moderate inhibition of P-gp | Gyémánt et al. (2006) | |
Fucoxanthin | Caco-2, CEM/ADR5000 | MTT assay, activity determination using Rh-123- and Calcein-AM retention assay | Significant enhancement of cytotoxicity and synergism using the following drugs: PTX, Cycloheximide, DOX, VBL, amphotericin-B, 5-FU, etoposide and cisplatine |
Eid et al. (2012) | |
Mutatochrome | L5178/MDR1 | Rh-123 accumulation assay | Moderate inhibition of P-gp | Gyémánt et al. (2006) | |
Retinoic acid | Caco-2, CEM/ADR5000 | MTT assay, activity determination using Rh-123- and Calcein-AM retention assay | Significant enhancement of cytotoxicity and synergism using the following drugs: DOX, VBL, 5-FU, etoposide and cisplatine | Eid et al. (2012) | |
Benzopyrones | |||||
Coumarins/coumaric acids | Auraptene | KB-C2 and KB/MRP | Accumulation assay of DNR in KB-C2 cells and calcein in KB/MRP cells, P-gp ATPase activity | Increase in the accumulation of DNR in KB-C2 cells, stimulation of the P-gp ATPase activity | Nabekura et al. (2008b) |
HT29 | Cytotoxicity assay (MTT), real-time RT-PCR | Synergic effects with cisplatin, DOX and VCR, increase in the toxicity of applied radiations in auraptene pretreated cells, overexpression of p21 in auraptene pretreated cells after radiotherapy | Moussavi et al. (2017) | ||
Bergamottin | K562/ADM | [3H]vincristine uptake | Increase in [3H]vincristine uptake, weak MDR reversal activity | Ikegawa et al. (2000) | |
Clausarin | K562/R7 | DNR accumulation assay | Inhibition of P-gp-mediated drug efflux | Bayet et al. (2007) | |
Dicynnamoyl-cis-khellactone | HepG2/Dox, K562/Dox | DOX accumulation and efflux assay, MDR reversing activity (cytotoxicity assay in the presence and absence of an anticancer drug), cell cycle distribution | Increase in DOX uptake and reduction of DOX efflux in HepG2/Dox cells, increase in cytotoxicity of anticancer drugs VBL, DOX, puromycin and PTX in HepG2/Dox and K562/Dox cells, enhancement of DOX-induced G2/M arrest in HepG2/Dox cells | Shen et al. (2006) | |
Dihydroxybergamottin | K562/ADM | [3H]vincristine uptake | Increase in [3H]vincristine uptake, weak MDR reversal activity | Ikegawa et al. (2000) | |
Phyllodulcin | KB-C2 | DNR accumulation | Increase in DNR accumulation (inhibition of P-gp-mediated efflux of DNR) | Nabekura et al. (2008a) | |
Praeruptorin A | HepG2/Dox, K562/Dox | MDR reversing activity (cytotoxicity assay in the presence and absence of an anticancer drug) | Increase in cytotoxicity of anticancer drugs VBL, DOX, puromycin and PTX in HepG2/Dox and K562/Dox cells | Shen et al. (2006) | |
Others | |||||
Dibenzopyran | Magniferin | Rats jejunal membrane in vitro | P-gp stimulation/inhibition profiles using a P-gp-dependent ATPase assay | Biphasic effect (stimulation at low concentration and inhibition at high concentration) | Najar et al. (2010) |
Phenols | [6]-Gingerol | KB-C2 | Determination of DNR and Rh-123 accumulation | Increase in accumulation of DNR and Rh-123 | Nabekura et al. (2005) |
Glucoside | Parishin C | Lymphoma cells | – | Moderate reduction of the activity of the efflux pump | Berek et al. (2001) |
Phenyl propanoid | Acteoside (Verbascosine) | Rats jejunal membrane in vitro | P-gp stimulation/inhibition profiles using a P-gp-dependent ATPase assay | Inhibitory effect on P-gp ATPase activity | Najar et al. (2010) |
*Saponins are marked with asterisks.
Research on olomoucine, purvalanol and roscovitine revealed amongst other things the inhibitive potential of purine alkaloids on P-gp in the form of MDCKII-ABCB1 cells and human HCT-8 and HepG2 cells while olomoucine had the strongest effect of all (Cihalova et al., 2013). Several groups have run experiments with the benzazepine alkaloid capsaicin on different cell lines such as KB-C2, Caco-2 and CEM/ADR5000. The most relevant result was the inhibition of P-gp efflux in the presence of digoxin as substrate at non-toxic concentrations (Nabekura et al., 2005; Han et al., 2006; Li et al., 2018a).
Isoflavonoids and some flavonoids often act as phytoestrogens. Investigations revealed that hydroxyl groups at position C-5 and C-7 are an important property for the P-gp inhibitory effect of flavonoids, although hydrophobicity usually promotes the affinity (Sheu et al., 2010). Acacetin, genistein, kaempferol and naringenin, were examined by Imai et al. (2004) and it resulted in positive effects on K562 cells expressing BCRP. The cytotoxicity of MTX and 7-ethyl-10-hydroxycamptothecin was enhanced using phytoestrogens. In addition, genistein and naringenin enhanced the accumulation of topotecan in K562/BCRP cells. Also apigenin had strong reversal effect on BCRP-mediated MDR (Imai et al., 2004). In MCF-7 MX100 cells, which also overexpressed BCRP, apigenin had similar effect on the accumulation of the anticancer agent MTX (Zhang et al., 2004). However, naringenin, which differs from apigenin only in the saturation of C-2 and C-3, had a significant loss of potency in comparison to the latter due to the lack of a double bond (Conseil et al., 1998). Several groups experimented with the isoflavone and phytoestrogen biochanin A and its outcome on BCRP and P-gp in cell lines such as Panc-1, MCF-7 MX100, MDA435/LCC6, MCF-7/ADR, MDA435/LCCMDR1 and Caco-2. In all studies, an inhibition of drug efflux, an increase of accumulation and a potentiation of cytotoxicity were observed. A combination of biochanin A with some other flavonoids yielded additive effects (Nguyen et al., 2003; Zhang and Morris, 2003; Zhang et al., 2004; Sergent et al., 2005). Nobiletin and tangeretin prevented the P-gp mediated efflux of [3H]vincristine (Ikegawa et al., 2000). Among other flavones with methoxyls they were examined by Ohtani et al. (2007) for the uptake potential of [3H]vincristine as a P-gp substrate. The MDR-reversing effect increased with the number of methoxyl moieties but with the exception of both at C-3′ and C-5′ position on B-ring. In this case there was even a decrease of MDR-reversing potency. Chrysin is a hydroxyflavone with simple structure which has also been examined by a number of groups. The competitive inhibition of the MRP-2 pumps in Caco-2 cells and an antiproliferative effect of chrysin are certain (Gyémánt et al., 2005; Sergent et al., 2005). Moreover, a prevention of BCRP and P-gp mediated efflux took place in MCF-7 MX100 cells and L5178 cells, respectively (Zhang et al., 2004; Zhang et al., 2005; Gyémánt et al., 2005). A prenyl or geranyl group at C-6 and C-8 of chrysin, and a prenyl in both positions ensured an improvement of the binding affinity to P-gp. In addition, there was more potency for inhibition after geranylation than prenylation which could be due to its level of lipophilicity (Di Pietro et al., 2002). In contrast, glycosylation taking place at any position tested, dramatically decreased the binding affinity of flavonoids (Sheu et al., 2010). Another way to gain binding affinity toward the C-terminal nucleotide-binding domain of P-gp, were other kinds of alkylation of chrysin including methyl, benzyl, isopropyl and 3,3-dimethylallyl (Boumendjel et al., 2002).
Several experiments have been carried out for the flavan epigallocatechin gallate (EGCG), a derivative of epigallocatechin (EGC) abundant in green tea. EGCG has been tested on different cells such as liver cancer cells Bel-7404/DOX, the colon cancer cell line Caco-2, leukemia cells CEM/ADR5000, endocervical adenocarcinoma cells KB-C2 and others. An increase of drug accumulation and sensitization in these and several other cell lines was observed (Kitagawa et al., 2004; Liang et al., 2010; Eid et al., 2013; Reyes-Esparza et al., 2015). Particularly substitutions at D-ring had positive effect on EGC and derivatives. A derivative of EGC with three methoxy groups in cis-configuration at D-ring and an oxycarbonylvinyl as its connection to C-3 led to a higher potency. This structure only regulated P-gp and could not affect BCRP or MRP1 (Wong et al., 2015).
While curcumin is a promising agent for liver protection and inhibition of cancerous cell growth, other curcuminoids like demethoxycurcumin and tetrahydrocurcumin have being studied more closely. They inhibited the efflux of chemptherapeutics and consequently increased cellular accumulation of drugs (Limtrakul et al., 2007; Ampasavate et al., 2010).
Among phenylpropanoids, some lignans (Figure 5) showed an inhibitory effect on P-gp. Schisandrin A was used on Caco-2 cells resulting in an increase in apical-to-basal transport and cytotoxicity Yoo et al. (2007b). A study which examined the structure activity of lignans showed the benefit of the absence of a hydroxyl group in position C-8 as in schisandrin A and γ-schizandrin for the function as p-gp inhibitor. In addition, a higher effect was seen in R-configurated biaryl than in the S-configuration (Slaninová et al., 2009). The reversal of cytotoxicity in CEM/ADR 5000 and Caco-2 cells was moreover successful by using menthol or thymol, monoterpenes obtained from volatile oils. They could reduce the IC50 value of doxorubicin which enhances the effectiveness of the drug (Eid et al., 2013).
Figure 5.
Chemical structures of some selected lignans with MDR reversal effects.
Reis et al. (2016) reported that jatrophane diterpenes including esulatin M, epoxywelwitschene, welwitschene, and euphowelwitschine A were more efficative in MDR cells than the positive control verapamil. In comparison to the known MDR modifier verapamil, euphoboetirane C, D, E, F, and G extracted from Euphorbia boetica showed multifold P-gp modulatory activity in L5178Y‐MDR cells (Neto et al., 2019). Three types of latilagascenes (G, H and I), macrocyclic diterpene esters, were effective in L5178 cells expressing P-gp as transport inhibitors (Duarte et al., 2008). All successfully tested diterpenes mentioned above had a cyclopentane moiety in their structure. An enormous increase of activity was observed due to their saturation. Moreover, there was lower MDR-reversing activity mostly when the number of three hydroxyls was exceeded, especially in position 5, 7, 9 and 12. Nevertheless hydropxyls at C-1, C-13, C-14 and C-15 had not any negative effect on their function (Zhu et al., 2016). Vasas et al. (2011) observed that the lack of oxygenation at C-2 increased the MDR-reversing activity in diterpene jatrophanes such as 3β,5α,15β-triacetoxy-7β-isobutanoyloxyjatropha-6(17),11E-diene-9,14-dione and 3β,5α,15β-triacetoxy-7β-isobutanoyloxy-9α-nicotinoyloxyjatropha-6(17),11-dien-14-one. Furthermore, they stated that the presence of benzoyl or nicotinoyl group at C-2 instead of an acetyl group increases the potency of jatrophane diterpenes. The reversal of MDR was noticed in some triterpenoids including ursolic acid tested in KB-C2 cells (Nabekura et al., 2010), and deacteylnomilin and limonin in CEM/ADR5000 cells (El-Readi et al., 2010). A study on triterpenoids such as tormentic acid and derivatives suggests that an acetylation of C-2 of a terpenoid can cause an increase of activity, while this is not the case with the acetylation of C-3 (da Graça Rocha et al., 2007). Carotenoids, a group derived from tetraterpenes, are substrates for ABC transporters and can effectively modulate MDR in cancer cells (Eid et al., 2012). A few types such as β-carotene (Eid et al., 2013) or the xanthophyll member capsorubin showed MDR reversal activity (Molnár et al., 2006). To mention another important group with aromatic members, coumarins and coumaric acids are benzopyrones which we presented in Tables 1 and 2. Auraptene for example not only caused an enhancement of drug uptake (Nabekura et al., 2008b); its effect even was of a synergistic kind with drugs such as cisplatin and VCR (Moussavi et al., 2017). Kasaian et al. (2015) examined fifteen sesquiterpene coumarins. Considering the structure-activity relationship they described that ring-opened drimane-type sesquiterpene coumarins such as lehmferin, farnesiferol B and farnesiferol C showed the best P-gp inhibitory effects. Their study also revealed that farnesiferol C inhibited the Rh-123 efflux even more than the positive control verapamil.
Table 2.
Phytochemicals Modulating Transporter or Protein Expression.
The effects of secondary metabolites on different cell lines expressing ABC-transporters – Regulation of the expression | |||||
---|---|---|---|---|---|
Substance | Cell line | Assay system | Result | Reference | |
Alkaloids | |||||
Quinolines, Isoquinolines, Quinazolines | Berbamine | K562/ADR | P-gp expression using flow cytometry, mdr-1 gene expression using RT-PCR | Reduction of MDR1 gene expression | Dong et al. (2004) |
Berberine | OC2 and KB (oral-), SC-M1 and NUGC-3 (gastric-), COLO 205 (colon cancer cell line) | Pgp-170 protein expression using flow cytometry | Upregulation of P-gp expression in tested cell lines, decrease in retention of Rh-123 | Lin et al. (1999a) | |
Hep3B, HepG2, HA22T/VGH |
Rh-123 retention and MDR1 transporter level using flow cytometry | Increase in MDR1 transporter level in HepG2, Hep3B, and HA22T/VGH cells, decrease in retention of Rh-123 in HepG2 | Lin et al. (1999b) | ||
A10 (rat vascular smooth muscle cells) | MDR1a and MDR1b gene expression by RT-PCR | Increase in expression of MDR1a and MDR1b mRNA | Suzuki et al. (2010) | ||
3Y1, dRLh-84, B16 | Rh-123 retention using flow cytometry, MDR1a and MDR1b protein expression using western blot | Increase in MDR1a and MDR1b protein level, decrease in intracellular Rh-123 concentration | Suzuki et al. (2010) | ||
Coptisine | A10 | MDR1a and MDR1b gene expression by RT-PCR, MDR1a and MDR1b protein expression using western blot, Rh-123 retention using flow cytometry | Increase in MDR1a and MDR1b mRNA and protein level, decrease in intracellular Rh-123 content | Suzuki et al. (2010) | |
3Y1, dRLh-84, B16 | Rh-123 retention using flow cytometry, MDR1a and MDR1b protein expression using western blot | Increase in the level of MDR1a and MDR1b protein, decrease in intracellular Rh-123 concentration | Suzuki et al. (2010) | ||
Fangchinoline | Caco-2, CEM/ADR5000 | MDR reversal assay (effect on DOX cytotoxicity), Rh-123 accumulation assay, western blot (P-gp expression level) | Increase in intracellular Rh-123 accumulation, decrease in P-gp expression, synergism in combination with DOX | Sun and Wink (2014) | |
Glaucine | MCF-7/ADR | MDR reversing activity, ADR and MTX efflux assay, real-time RT-PCR, P-gp and MRP1 ATPase activity assay | Inhibition of P-gp and MRP1-mediated efflux, suppression of the expression of MDR1 and MRP1 genes, reversion of the resistance of MCF-7/ADR to ADR and MTX, increase in P-gp and MRP1 ATPase activities | Lei et al. (2013) | |
O-(4-ethoxyl-butyl)-berbamine | MCF-7/ADR | MCF-7/ADR sensitivity to ADR (MTT assay), Rh-123 retention using flow cytometry, expression of mdr-1gene using RT-PCR | Sensitization to ADR, increase in the accumulation of Rh-123, reduction of MDR1 gene expression | Cheng et al. (2006) | |
Palmatine | A10 | MDR1a and MDR1b gene expression by RT-PCR | Increase in the expression of MDR1a and MDR1b mRNA | Suzuki et al. (2010) | |
3Y1, dRLh-84, B16 | MDR1a and MDR1b protein expression using western blot, Rh-123 retention using flow cytometry | Increase in the level of MDR1a and MDR1b protein, increase in intracellular Rh-123 content | Suzuki et al. (2010) | ||
Tetrandrine | Caco-2, CEM/ADR5000 | MDR reversal assay (effect on DOX cytotoxicity), Rh-123 accumulation assay, western blot (P-gp expression level) | Increase in intracellular Rh-123 accumulation, decrease in P-gp expression, synergism in combination with DOX | Sun and Wink (2014) | |
K562 (DOX-treated vs. tetrandrine+DOX-treated group) | mRNA expression using RT-PCR, detecting P-gp expression as well as Rh-123 accumulation by FACS | Decrease in DOX-induced MDR1 mRNA expression, inhibition of DOX-induced P-gp expression, increase in Rh-123 accumulation | Shen et al. (2010) | ||
Quinolizidine alkaloids | Matrine | MCF-7/ADR | Western blot for labelling P-gp and MRP1 proteins, fluorospectrophotometry for ADR accumulation assay | Reduction of P-gp expression, increase in intracellular accumulation of ADR, decrease in cell growth | Zhou et al. (2017) |
Indoles and β-carbolines | Antofine | A549-PA | P-gp expression using western blot, MDR-1 mRNA expression using RT-PCR, Rh-123 accumulation by FACS | Reduction of P-gp and MDR-1 mRNA expression, increase in intracellular Rh-123 content, synergism with PTX | Kim et al. (2012) |
Ephedrine | K562/A02 | MDR1 gene expression using semi-quantitative RT-PCR, P-gp expression using western blot | Decrease in MDR1 mRNA and P-gp expression | Gao et al. (2008) | |
Indole-3-carbinol | Hepatocytes of mouse treated with combination of indole-3-carbinol and VBL/VCR (in vivo) | P-gp expression using western blot, quantitative stereology using immunohistochemical staining with anti-P-gp antibody | Inhibition of VBL/VCR-induced P-gp expression | Arora and Shukla (2003) | |
Staurosporine | MDR KB-V1 | Cytotoxicity, P-gp expression using western blot, MDR1 gene expression by northern blot | Cytotoxicity synergism with verapamil, sensitization of cells to VBL when co-treated with verapamil, decrease in P-gp and MDR1 gene expression | Sampson et al. (1993) | |
Vauqueline | K562/A02 | MDR1 gene expression using semi-quantitative RT-PCR, P-gp expression using western blot | Decrease in MDR1 mRNA and P-gp expression | Gao et al. (2008) | |
Piperidines, Pyrazines, Diketopiperazines | Piperine | MCF-7/Dox | ABCB1 and ABCG2 mRNA expression by semi-quantitative RT-PCR; fluorescent dye efflux assay, cytotoxicity assay (MTT) | Decrease in ABCB1 and ABCG2 mRNA expression, increase in intracellular Rh-123 and MTX accumulation, reversal of resistance to DOX and MTX | Li et al. (2011) |
A-549/DDP | ABCC1 mRNA expression by semi-quantitative RT-PCR; fluorescent dye efflux assay, cytotoxicity assay (MTT) | Decrease in ABCC1 mRNA expression, increase in intracellular DOX accumulation, reversal of resistance to DOX | |||
Tetramethylpyrazine | MCF-7/Dox | DOX retention by flow cytometry, P-gp expression using western blot, MDR1 expression using RT-PCR | Increase in DOX retention, reversal of resistance to PTX, VCR and DOX; decrease in P-gp and MDR1 mRNA expression | Zhang et al. (2012) | |
Pumc-91/ADM, T24/DDP |
Cell viability assay using Cell Counting Kit-8, qRT-PCR for MRP1 mRNA, western blot and immunofluorescence assay for MRP1 protein expression | Decrease in MRP1 protein and mRNA expression, reversal of MDR (increase in cytotoxicity of ADR and DDP) | Wang et al. (2016) | ||
BEL-7402/ADM | ADR accumulation by flow cytometry and HPLC, gene expression using RT-PCR, protein expression by western blot | Decrease in MDR1, MRP2, MRP3 and MRP5 mRNA and proteins expression, increase in ADR intracellular accumulation, increase in ADR cytotoxicity (MDR reversal effect) | Wang et al. (2010) | ||
Acridone alkaloids | Gravacridonetriol | L5178/MDR1 | Rh-123 retention, MTT antiproliferative assay, MDR1 expression by RT-PCR | Decrease in Rh-123 efflux, increase in DOX cytotoxicity (synergism), decrease in MDR1 mRNA expression | Rethy et al. (2008) |
Gravacridonediol monomethyl ether | L5178/MDR1 | Rh-123 retention, MTT antiproliferative assay, MDR1 expression by RT-PCR | Decrease in Rh-123 efflux, increase in DOX cytotoxicity (synergism), decrease in MDR1 mRNA expression | Rethy et al. (2008) | |
Nucleosides | Clitocine | R-HepG2, MES-SA/Dx5 | DOX retention by flow cytometry, western blot for P-gp and qRT-PCR for MDR1 expression | Decrease in MDR1 mRNA and P-gp expression, reversal of MDR (enhancement of DOX cytotoxicity), increase in DOX accumulation | Sun et al. (2012) |
Sulfinosine | NCI-H460/R, U87-TxR | DOX retention by flow cytometry, western blot for P-gp and RT-PCR for MDR1 mRNA expression | Decrease in MDR1 mRNA and P-g expression, increase in DOX accumulation, decrease in resistance to DOX (enhancement of DOX cytotoxicity) | Dačević et al. (2013) | |
Further alkaloids | Capsaicin | Caco-2 | [3H]-digoxin retention, western blot for P-gp and RT-PCR for MDR1 mRNA expression | Increase in P-gp and MDR1 mRNA expression, increase in [3H]-digoxin efflux | Han et al. (2006) |
Homoharringtonine | CEM/E1000 | MTT assay | Decrease in cell viability, modulation of MRP1-mediated MDR | Efferth et al. (2002) | |
Polyphenols | |||||
Flavonoids | Ampelopsin | K562/ADR | MTT assay, P-gp expression by PE-labeled antibody, ADR accumulation using flow cytometry | Decrease in P-gp expression, increase in ADR cytotoxicity and intracellular accumulation, synergism with ADR | Ye et al. (2009) |
Baicalein | LS174T, HepG2 | MTT assay, MDR1 expression using real-time PCR | Decrease in cell viability, increase in MDR1 expression in LS174T | Li et al. (2010) | |
Epicatechingallate | Bel-7404/DOX | MTT assay, Rh-123 retention by flow cytometry, intracellular DOX content using fluorospectrophotometry, semi-quantitative RT-PCR, P-gp expression by FACS using anti-P-gp monoclonal antibody | Increase in DOX and Rh-123 accumulation, and DOX cytotoxicity (synergism), decrease in MDR1 and P-gp expression | Liang et al. (2010) | |
Epigallocatechin gallate | Bel-7404/DOX | MTT assay, Rh-123 retention by flow cytometry, intracellular DOX content using fluorospectrophotometry, semi-quantitative RT-PCR, P-gp expression by FACS using anti-P-gp monoclonal antibody | Increase in DOX and Rh-123 accumulation, and DOX cytotoxicity (synergism),decrease in MDR1 and P-gp expression | Liang et al. 2010) | |
MCF-7Tam | MTT assay, RT-PCR, western blot for P-gp and BCRP protein expression, Rh-123 and MTX accumulation | Reduction of cell proliferation, decrease in P-gp and BCRP expression, increase in MTX accumulation | Farabegoli et al. (2010) | ||
MCF-7 | Cytotoxicity using MTS assay, P-gp protein expression using western blot and immunofluorescence microscopy, Rh-123 accumulation | Increase in Rh-123 accumulation, decrease in P-gp protein expression, reduction of cell viability | Reyes-Esparza et al. (2015) | ||
Quercetin | HL-60/ADM, K562/ADM | MDR reversal by MTT assay, RT-PCR, flow cytometry | Sensitization to DNR, decrease in MRP1 gene and protein expression | Cai et al. (2004; 2005) | |
BEL/5-FU | Cytotoxicity assay; Rh-123 and ADR accumulation using flow cytometry; ABCB1, ABCC1, ABCC2 mRNA and protein expression using real-time PCR and western blot | Increase in sensitivity to chemotherapeutic drugs 5-FU, MMC and ADR; increase in Rh-123 and ADR accumulation; decrease in ABCB1, ABCC1 and ABCC2 mRNAs and proteins expression | Chen et al. (2018) | ||
Curcuminoids | Bisdemethoxycurcumin | KB-V1 | RT-PCR, western blot | Decrease in MDR1 and P-gp expression | Limtrakul et al. (2004) |
Curcumin | SKOV3(TR) | Cytotoxicity in combination with PTX, western blot | Enhancement of PTX cytotoxic activity, reduction of P-gp expression | Ganta and Amiji (2009) | |
HCT-8/VCR | MTT assay, Rh-123 retention, MDR1 and survivin genes expression by RT-PCR | Reversal of MDR, decrease in MDR1 expression, increase in substrate accumulation | Lu et al. (2013) | ||
Neo-/lignans | Honokiol | MCF-7/ADR | MTT assay, Rh-123 retention, qPCR, P-gp expression by FACS | Increase in Rh-123 retention, reduction of MDR1 and P-gp expression, increase in ADR cytotoxicity | Xu et al. (2006) |
Dibenzocyclo-octadienelignans | Schisandrin A (Deoxyschizandrin) |
KBV200, MCF-7/Dox, Bel7402 | MTT assay (cytotoxicity in combination with DOX, VCR and PTX), DOX and Rh-123 retention, RT-PCR, western blot for P-gp expression | Increase in Dox and Rh-123 retention, reduction of MDR1 and P-gp expression, sensitization to cytotoxic drugs | Huang et al. (2008) |
COR-L23/R | MTT assay, DOX retention by flow cytometry | Elevation of DOX cytotoxicity, increase in DOX accumulation | Slaninová et al. (2009) | ||
Schisandrin B/γ-Schizandrin | COR-L23/R | MTT assay, DOX retention by flow cytometry | Elevation of DOX cytotoxicity, increase in DOX accumulation | Slaninová et al. (2009) | |
Terpenoids | |||||
Sesquiterpenes | Artesunate | CEM/E1000, CEM/VLB100 |
MTT assay, DNR retention assay by flow cytometry | Decrease in cell viability, increase in DNR accumulation, modulation of MDR1- and MRP1-mediated MDR | Efferth et al. (2002) |
EGb761 | HepG2 | MDR1 expression by real-time RT-PCR | Induction of MDR1 expression | Li et al. (2009) | |
Diterpenes | Ginkgolide A and B | HepG2 | MDR1 expression by real-time RT-PCR | Induction of MDR1 expression | Li et al. (2009) |
Triptolide | In vitro (KB-7D, KB-tax) In vivo (KB-7D-,KB-tax-bearing mouse) |
In vitro growth inhibition assay, western blot for MDR1 and MRP1 protein expression, in vivo tumor weight evaluation | Inhibition of cell growth, decrease in MDR1 and MRP1 protein expression, inhibition of tumor growth, decrease in tumor weight when combined with 5-fluorouracil (synergism effect) | Chen et al. (2010) | |
DU145/ADM | MTT assay, RT-PCR, western blot | Increase in ADR cytotoxicity, reduction of MDR1 mRNA and protein expression | Guo et al. (2013) | ||
Benzopyrones | |||||
Coumarins/coumaric acids | Pyranocoumarins | KB-V1 | Sulforhodamine B cytotoxicity assay, DOX retention by flow cytometry, MDR1 mRNA Expression using RT-PCR, P-gp expression by western blot | Synergism with DOX, VCR, puromycin and PTX; increase in intracellular accumulation of DOX; decrease in P-gp and MDR1 mRNA expression | Wu et al. (2003) |
Phenols | |||||
6-Gingerol, 10-Gingerol | PC3R | Cytotoxicity assay, MRP-1 protein expression using western blot | Reduction of cell survival, decrease in MRP-1 protein expression | Liu et al. (2017) | |
6-Shogaol, 10-Shogaol | PC3R | Cytotoxicity assay, MRP-1 protein expression using western blot | Reduction of cell survival, decrease in MRP-1 protein expression | Liu et al. (2017) |
The studies introduced phytochemicals with completely different basic structures, each showing both strong and ineffective members in targeting ABC-transporters. The molecules mentioned were a selection chosen due to their effectiveness, especially in relation to the substances to which they were compared.
Phytochemicals Modulating Transporter or Protein Expression
The following discussion focusses on the question how far phytochemicals affect the expression of ABC transporters or proteins in cancer cell lines. A list of relevant publications is documented in Table 2. Although many investigations addressed the inhibitory effect of secondary metabolites on transporter activity, many phytochemicals regulate the expression of corresponding genes, including alkaloids, polyphenols, lignans, terpenes and benzopyrones. Berbamine caused a downregulation of Mdr-1 expression in K562/ADR cells (Dong et al., 2004), and so did its derivative O-(4-ethoxylbutyl)-berbamine (Cheng et al., 2006). Glaucine not only functioned as inhibitor of transporters but also reduced the expression of MDR1 and MRP1 (Lei et al., 2013) while the isoquinoline alkaloids berberine and coptisine, mediated the expression of P-gp (Lin et al., 1999a). Tetramethylpyrazine downregulated the expression of MDR1, MRP2, MRP3 and MRP5 in BEL-7402/ADM cells (Wang et al., 2010). It has also reduced the expression of MDR1 and MRP1 in MCF-7/Dox and Pumc-91/ADM cells, respectively (Zhang et al., 2012; Wang et al., 2016). MDR1 mRNA and P-gp expression were reduced in response to clitocine and sulfinosine in several cell lines, such as R-HepG2, MES-SA/Dx5, NCI-H460/R and U87-TxR (Sun et al., 2012; Dačević et al., 2013). Among polyphenols, EGCG mediated a reduction of P-gp and BCRP expression resulting in drug accumulation (Farabegoli et al., 2010). Curcumin enhanced the cytotoxicity of PTX and ADR, respectively against SKOV3(TR) and K562/A02 cells by reducing the expression of P-gp in both cell lines (Chang et al., 2006; Ganta and Amiji, 2009). While some diterpenes and sesquiterpenes induced the transporter expression (Li et al., 2009), others like triptolide suppressed the expression of MDR1 and MRP1 proteins in KB-7D and KB-tax cells (Chen et al., 2010). Beside the positive effect of some coumarins on the activity of ABC transporters, pyranocoumarin was tested on KB-V1 cells and showed a reduction of P-gp protein and MDR1 expression (Wu et al., 2003).
Athough no clear relationship could be established between the structure of a modulator with thre expression of a transporter protein, these findings can be relevant for clinical applications.
Synergistic Combinations of Chemotherapeutics With Phytochemicals
In addition to affecting the activity or expression of transporters, phytochemicals can also reverse MDR in cancer cells through synergism. Phytochemicals causing synergistic interactions with anticancer drugs are documented in Tables 1 and 2. The two-drug combination of harmine with DOX not only showed an enhancement of cytotoxicity but a synergistic effect. Glaucine potentiated DOX toxicity in Caco-2 and CEM/ADR5000 cells and reversed their resistance to anticancer drugs. Glaucine exerted synergistic interaction with DOX and even more in a three drug combination with DTN (Eid et al., 2013). Euphoboetirane C, D, F, G, H and I extracted from Euphorbia boetica showed strong synergistic interactions with DOX in L5178Y‐MDR cells (Neto et al., 2019). Nobiletin and antofine showed synergistic interactions with PTX (Kim et al., 2012; Ma et al., 2015). Jatrophane diterpenes including euphowelwitschine A, euphowelwitschine B, epoxywelwitschene, esulatin M isolated from Euphorbia welwitschii demonstrated synergism with DOX in L5178Y‐MDR cells (Reis et al., 2016). Fangchinoline, tetrandrine, and pyranocoumarins has been shown to synergistically increase the cytotoxicity of DOX (Wu et al., 2003; Sun and Wink, 2014). These synergistic effects are probably caused by interference of the phytochemicals with ABC transporters and additional molecular targets in cancer cells (Wink et al., 2012). These studies show that a combination of substances can have a great advantage over using single drugs against cancer cells by exploiting synergism.
In Silico Analysis of Phytochemicals as MDR Reversing Agents
In Silico experiments are promising methods for making predictions about interactions of molecules to the drug-binding site of a target such as P-gp, BCRP and MRP1. Molecular docking studies can help finding the phytochemicals with best affinity for the drug-binding site which should be used as lead molecules for further studies. The nucleotide-binding domains (NBD) of these ABC-Transporters are hydrophilic protein parts (Jones and George, 2002). In addition to essential hydrogen bonds which play a crucial role in maintaining the stability and function of biomolecules, modulators with hydrophobic moieties often show affinties to NBDs.
The benzolisoquinoline alkaloid tetrandrine was confirmed to have inhibitory capability. Its binding affinity is close to verapamil and their docking positions are similar. Main part of tetrandrine is fixed in a lipophilic pocket formed by the amino acids Ala729, Ala987, Ileu306, Ileu340, Leu339, Leu65, Leu975, Met69, Met986, Phe303, Phe336, Phe343, Phe728, Phe732, Phe983, Tyr307, Tyr310. Moreover, the positively charged methylamine moiety of tetrandrine formed a cation-π interaction with Phe343 and an aryl ether got into a π-π interaction with Phe336 (Liao et al., 2019).
Regarding in silico investigations, another subtype of alkaloids, piperine was examined by Syed et al. (2017) revealing that hydrophobic interactions with P-gp took place in following positions: Leu339, Met69, Met986, Phe72, Phe336, Phe728, Phe983, Tyr953 and Val982. Moreover, an H-bond with Tyr307 was established. Two piperine analogs were designed which showed better hydrophobic interaction with most of the amino acids mentioned (Syed et al., 2017).
Pharmacophore modeling which was carried out for acridones indicated that three aromatic rings and two H–acceptors which are given in position C-9 bearing a carbonyl group and N-10 were conducive for stable docking. Including designed analogs of acridone the oxygen of a morpholine moiety at a phenyl ring built a water bridge with Ser309 of P-gp and a carboxamide at C-4 helped docking with Phe343. This analog showed hydrophobic interactions with Ala229, Ala302, Ala342, Ile218, Ile221, Ile299, Ile306, Leu225 with even more than two interactions, Leu339, Phe343, and Val345 of P-gp (Gade et al., 2018).
Badhan and Penny (2006) worked on the main characteristics of the pharmacophore of flavonoids by in silico modeling. Flavonoids tested get located within the C-terminal NBD of P-gp binding to amino acids of the ATP pocket such as Tyr1044. The hydroxyl moiety at C-5 of natural flavonoids can form a water bridge with Lys1076 while the hydroxyl group in position 3 and the carbonyl group get into hydrogen bonds within the NBD. Hydroxyl groups at position C-7 led to important hydrogen bonds with amino acids for several flavonoids such as EGC and chrysin (Wongrattanakamon et al., 2017). Ring B gets involved in π- π interaction with Tyr1044. If benzyl, geranyl or prenyl groups added at C-6 and several substitutes added at C-8 position, the docking capability was enhanced. Molecular docking analysis demonstrated that apigenin binds to the ATP-binding site of P-gp through a hydrogen bond with LYS408 and therefore, interferes with binding and cleavage of ATP which are vital for the function of ABC transporters (Saeed et al., 2015). Comparing flavonols to their corresponding flavones such as kaempferol to apigenin, better docking properties have been found in flavonols due to the number of hydrogen bonds (Badhan and Penny, 2006). A number of naringenin derivatives such as hydrazones and azines were produced by Ferreira et al. (2018) to achieve an improved MDR reversal in P-gp and BCRP. There was a high selectivity for most members of these two groups. It has been shown that the active compounds bind to hydrophobic residues such as transmembrane helix 4 and 5 in the center of an active pocket of BCRP similar to fumitremorgin C, a common inhibitor of BCRP. The hydrazone of these derivatives, a continuation of imine, is located in a pocket in π- π interaction with Phe515 and a dipole-dipole interaction with Met541. The B-ring is in another lipophilic bag made of Phe545, Pro574 and Val516. An improvement in the interaction between the transporter MRP1 and the carbohydrazide derivatives of flavonones due to the resulting flexibility was also observed (Ferreira et al., 2018).
Conclusions and Future Perspectives
Due to the increase of cancer cases and restrictions in therapy owing to the degree of harmfulness, effectiveness and the associated high costs, the development of new substances with less side effects and higher efficacy is required. So far, many structures from the plant kingdom have been discovered and used, such as, diterpene derivatives as taxanes and vinca alkaloids are prime examples with regard to antitumor therapy. Therefore, numerous secondary metabolites which already had positive effect on cell lines should be taken into account for further investigations, which is why we have tried to list them extensively in our tables.
Many phytochemicals including alkaloids, flavonoids, curcuminoids, stilbenoids, terpenes, carotenoids, lignans, and polyketides were examined for their pharmacological activity against MDR. In most of the studies it was possible to differentiate between more effective and less effective molecules. As the next step these molecules must be further studied to determine molecular mechanisms and to identify the pharmacophores. Some valuable results have been provided by structure-activity relationship studies or in silico modeling as shown in this article. Subtances which were superior to others should be selected for further research, so possible lead molecules can be developed. A further step to increase efficacy could be the use of new drug delivery methods and controlled release as with nanotechnologies. As shown in this review, the combination of two and more modulators of differing structures and mode of actions together with chemotherapeutics is another interesting approach. The resulting additive, but above all synergistic effects can be of great importance. They would allow reducing the dose of chemotherapeutics resulting in less side effects and a higher compliance of patients.
Summin up, many phytochemicals in the group of alkaloids, flavonoids, curcuminoids, stilbenoids, terpenes, carotenoids, lignans, and polyketides have been investigated for MDR-reversing activity. Phytochemicals can be a promising source of adjuvant chemicals against cancer, not at least because of their generally low cytotoxicity in the human body. The adjuvant uses of MDR reversing phytochemicals in combination with anticancer drugs may improve the treatment of multidrug resistant cancer types. The present review summarized reports of several secondary metabolites that are capable of synergistically reversing MDR and inhibiting chemotherapy-resistant cancer cells by affecting transporter activity and the expression of ABC transporter genes. Synergism would allow reducing the dose of chemotherapeutics resulting in less side effects and a higher compliance of patients. The efficacy of phytochemicals needs to be confirmed clinically, but nevertheless they already can be considered as the fourth generation of ABC transporter modulators.
Author Contributions
BT and IS performed the literature search and wrote the first draft of the manuscript. MW revised and edited the manuscript. All the authors approved the final version of the manuscript.
Funding
Ruprecht-Karls-Universität Heidelberg provided financial support within Open-Access Publishing Program.
Conflict of Interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Abbreviations
ADR, adriamycin; CPT, camptothecin; DOX, doxorubicin; DNR, daunorubicin; DTN, digitonin; EGC, epigallocatechin; EGCG, epigallocatechin gallate; MDR, multidrug resistance; MTX, mitoxantrone; P-gp, P-glycoprotein; PTX, paclitaxel; Rh-123, rhodamine 123; VBL, vinblastine; VCR, vincristine; BCRP, breast cancer resistance protein; MRP, multidrug resistance protein.
References
- Aktories K., Förstermann U., Hofmann F. B., Starke K. (2017). Allgemeine und spezielle Pharmakologie und Toxikologie: Begründet von W. Forth. Eds. Henschler D., Rummel W. (München, Germany: Urban & Fischer Verlag/Elsevier GmbH; ). [Google Scholar]
- Alamolhodaei N. S., Tsatsakis A. M., Ramezani M., Hayes A. W., Karimi G. (2017). Resveratrol as MDR reversion molecule in breast cancer: An overview. Food Chem. Toxicol. 103, 223–232. 10.1016/j.fct.2017.03.024 [DOI] [PubMed] [Google Scholar]
- Ampasavate C., Sotanaphun U., Phattanawasin P., Piyapolrungroj N. (2010). Effects of Curcuma spp. on P-glycoprotein function. Phytomedicine 17, 506–512. 10.1016/j.phymed.2009.09.004 [DOI] [PubMed] [Google Scholar]
- Arora A., Shukla Y. (2003). Modulation of vinca-alkaloid induced P-glycoprotein expression by indole-3-carbinol. Canc. Lett. 189, 167–173. 10.1016/S0304-3835(02)00550-5 [DOI] [PubMed] [Google Scholar]
- Badhan R., Penny J. (2006). In silico modelling of the interaction of flavonoids with human P-glycoprotein nucleotide-binding domain. Eur. J. Med. Chem. 41, 285–295. 10.1016/j.ejmech.2005.11.012 [DOI] [PubMed] [Google Scholar]
- Baumann T. W., Frischknecht P. M. (1988). “Purines,” in Phytochemicals in Plant Cell Cultures (San Diego, USA: Academic Press; ), 403–417. [Google Scholar]
- Bayet C., Fazio C., Darbour N., Berger O., Raad I., Chaboud A., et al. (2007). Modulation of P-glycoprotein activity by acridones and coumarins from Citrus sinensis. Phytother. Res. 21, 386–390. 10.1002/ptr.2081 [DOI] [PubMed] [Google Scholar]
- Berek L., Szabo D., Petri I. B., Shoyama Y., Lin Y. H., Molnár J. (2001). Effects of naturally occurring glucosides, solasodine glucosides, ginsenosides and parishin derivatives on multidrug resistance of lymphoma cells and leukocyte functions. Vivo 15, 151–156. [PubMed] [Google Scholar]
- Bernstein C., Bernstein H., Payne C. M., Garewal H. (2002). DNA repair/pro-apoptotic dual-role proteins in five major DNA repair pathways: fail-safe protection against carcinogenesis. Mutat. Res. Rev. Mutat. 511, 145–178. 10.1016/S1383-5742(02)00009-1 [DOI] [PubMed] [Google Scholar]
- Bloch K. E. (1983). Sterol, structure and membrane function. Crit. Rev. Biochem. 14, 47–92. 10.3109/10409238309102790 [DOI] [PubMed] [Google Scholar]
- Borst P., Evers R., Kool M., Wijnholds J. (2000). A family of drug transporters: the multidrug resistance-associated proteins. J. Natl. Canc. Inst. 92, 1295–1302. 10.1093/jnci/92.16.1295 [DOI] [PubMed] [Google Scholar]
- Borthwick A. D., Liddle J. (2011). The design of orally bioavailable 2, 5-diketopiperazine oxytocin antagonists: from concept to clinical candidate for premature labor. Med. Res. Rev. 31, 576–604. 10.1002/med.20193 [DOI] [PubMed] [Google Scholar]
- Boumendjel A., Di Pietro A., Dumontet C., Barron D. (2002). Recent advances in the discovery of flavonoids and analogs with high-affinity binding to P-glycoprotein responsible for cancer cell multidrug resistance. Med. Res. Rev. 22, 512–529. 10.1002/med.10015 [DOI] [PubMed] [Google Scholar]
- Brown G. A., McPherson J. P., Gu L., Hedley D. W., Toso R., Deuchars K. L., et al. (1995). Relationship of DNA topoisomerase IIα and β expression to cytotoxicity of antineoplastic agents in human acute lymphoblastic leukemia cell lines. Canc. Res. 55, 78–82. [PubMed] [Google Scholar]
- Cai X., Chen F. Y., Han J. Y., Gu C. H., Zhong H., Ouyang R. R. (2004). Restorative effect of quercetin on subcellular distribution of daunorubicin in multidrug resistant leukemia cell lines K562/ADM and HL-60/ADM. Chin. J. Canc. 23, 1611–1615. [PubMed] [Google Scholar]
- Cai X., Chen F. Y., Han J. Y., Gu C. H., Zhong H., Teng Y., et al. (2005). Reversal of multidrug resistance of HL-60 adriamycin resistant leukemia cell line by quercetin and its mechanisms. Chin. J. Oncol. 27, 326–329. [PubMed] [Google Scholar]
- Cao J., Chen X., Liang J., Yu X. Q., Xu A. L., Chan E., et al. (2007). Role of P-glycoprotein in the intestinal absorption of glabridin, an active flavonoid from the root of Glycyrrhiza glabra. Drug Metabol. Dispos. 35, 539–553. 10.1124/dmd.106.010801 [DOI] [PubMed] [Google Scholar]
- Cao Z., Wright M., Cheng J., Huang X., Liu L., Wu L., et al. (2014). The novel bis-benzylisoquinoline PY35 reverses P-glycoprotein-mediated multidrug resistance. Oncol. Rep. 32, 1211–1217. 10.3892/or.2014.3326 [DOI] [PubMed] [Google Scholar]
- Chanmahasathien W., Ampasavate C., Greger H., Limtrakul P. (2011). Stemona alkaloids, from traditional Thai medicine, increase chemosensitivity via P-glycoprotein-mediated multidrug resistance. Phytomedicine 18, 199–204. 10.1016/j.phymed.2010.07.014 [DOI] [PubMed] [Google Scholar]
- Chang H. Y., Pan K. L., Ma F. C., Jiao X. Y., Zhu H. F., Liu J. H., et al. (2006). The Study on Reversing Mechanism of Multidrug Resistance of K562/A02 Cell Line by Curcumin and Erythromycin. Chin. J. Hematol. 27, 254–258. [PubMed] [Google Scholar]
- Chen Y. W., Lin G. J., Chuang Y. P., Chia W. T., Hueng D. Y., Lin C. K., et al. (2010). Triptolide circumvents drug-resistant effect and enhances 5-fluorouracil antitumor effect on KB cells. Anti Canc. Drugs 21, 502–513. 10.1097/CAD.0b013e328337337c [DOI] [PubMed] [Google Scholar]
- Chen Z., Shi T., Zhang L., Zhu P., Deng M., Huang C., et al. (2016). Mammalian drug efflux transporters of the ATP binding cassette (ABC) family in multidrug resistance: A review of the past decade. Canc. Lett. 370, 153–164. 10.1016/j.canlet.2015.10.010 [DOI] [PubMed] [Google Scholar]
- Chen Z., Huang C., Ma T., Jiang L., Tang L., Shi T., et al. (2018). Reversal effect of quercetin on multidrug resistance via FZD7/β-catenin pathway in hepatocellular carcinoma cells. Phytomedicine 43, 37–45. 10.1016/j.phymed.2018.03.040 [DOI] [PubMed] [Google Scholar]
- Cheng Y. H., Qi J., Xiong D. S., Liu J. W., Qi S. L., Pan B., et al. (2006). Reversal of multidrug resistance in drug-resistant human breast cancer cell line MCF-7/ADR by calmodulin antagonist O-(4-ethoxyl-butyl)-berbamine. Acta Academiae Medicinae Sinicae 28, 164–168. [PubMed] [Google Scholar]
- Choi C. H., Kang G., Min Y. D. (2003). Reversal of P-glycoprotein-mediated multidrug resistance by protopanaxatriol ginsenosides from Korean red ginseng. Planta Med. 69, 235–240. 10.1055/s-2003-38483 [DOI] [PubMed] [Google Scholar]
- Chung S. Y., Sung M. K., Kim N. H., Jang J. O., Go E. J., Lee H. J. (2005). Inhibition of P-glycoprotein by natural products in human breast cancer cells. Arch. Pharmacal Res. 28, 823–828. 10.1007/BF02977349 [DOI] [PubMed] [Google Scholar]
- Cihalova D., Hofman J., Ceckova M., Staud F. (2013). Purvalanol A, olomoucine II and roscovitine inhibit ABCB1 transporter and synergistically potentiate cytotoxic effects of daunorubicin in vitro. PloS One 8, e83467. 10.1371/journal.pone.0083467 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coley H. M. (2008). Mechanisms and strategies to overcome chemotherapy resistance in metastatic breast cancer. Canc. Treat. Rev. 34, 378–390. 10.1016/j.ctrv.2008.01.007 [DOI] [PubMed] [Google Scholar]
- Collin G., Höke H. (2000). “Quinoline and isoquinoline,” in Ullmann"s Encyclopedia of Industrial Chemistry (New Jersey, USA: Wiley-VCH; ). [Google Scholar]
- Conseil G., Baubichon-Cortay H., Dayan G., Jault J. M., Barron D., Di Pietro A. (1998). Flavonoids: a class of modulators with bifunctional interactions at vicinal ATP-and steroid-binding sites on mouse P-glycoprotein. Proc. Natl. Acad. Sci. 95, 9831–9836. 10.1073/pnas.95.17.9831 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cör D., Knez Ž., Knez Hrnčič M. (2018). Antitumour, antimicrobial, antioxidant and antiacetylcholinesterase effect of Ganoderma lucidum terpenoids and polysaccharides: A review. Molecules 23, 649. 10.3390/molecules23030649 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cripe L. D., Uno H., Paietta E. M., Litzow M. R., Ketterling R. P., Bennett J. M., et al. (2010). Zosuquidar, a novel modulator of P-glycoprotein, does not improve the outcome of older patients with newly diagnosed acute myeloid leukemia: a randomized, placebo-controlled trial of the Eastern Cooperative Oncology Group 3999. Blood 116, 4077–4085. 10.1182/blood-2010-04-277269 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cseke L. J., Kirakosyan A., Kaufman P. B., Warber S. L., Duke J. A., Brielmann H. L. (2006). Natural Products from Plants. 2nd Ed (Boca Raton, USA: CRC Press; ), ISBN: 0-8493-2976-0. [Google Scholar]
- da Graça Rocha G., Simoes M., Lucio K. A., Oliveira R. R., Kaplan M. A. C., Gattass C. R. (2007). Natural triterpenoids from Cecropia lyratiloba are cytotoxic to both sensitive and multidrug resistant leukemia cell lines. Bioorg. Med. Chem. 15, 7355–7360. 10.1016/j.bmc.2007.07.020 [DOI] [PubMed] [Google Scholar]
- Dačević M., Isaković A., Podolski-Renić A., Isaković A. M., Stanković T., Milošević Z., et al. (2013). Purine nucleoside analog-sulfinosine modulates diverse mechanisms of cancer progression in multi-drug resistant cancer cell lines. PloS One 8, e54044. 10.1371/journal.pone.0054044 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Daenen S., van der Holt B., Verhoef G. E., Löwenberg B., Wijermans P. W., Huijgens P. C., et al. (2004). Addition of cyclosporin A to the combination of mitoxantrone and etoposide to overcome resistance to chemotherapy in refractory or relapsing acute myeloid leukaemia: A randomised phase II trial from HOVON, the Dutch–Belgian Haemato-Oncology Working Group for adults. Leuk. Res. 28, 1057–1067. 10.1016/j.leukres.2004.03.001 [DOI] [PubMed] [Google Scholar]
- De Castro W. V., Mertens-Talcott S., Derendorf H., Butterweck V. (2008). Effect of grapefruit juice, naringin, naringenin, and bergamottin on the intestinal carrier-mediated transport of talinolol in rats. J. Agr. Food Chem. 56, 4840–4845. 10.1021/jf0728451 [DOI] [PubMed] [Google Scholar]
- de Sa A., Fernando R., Barreiro E. J., Fraga M., Alberto C. (2009). From nature to drug discovery: the indole scaffold as a ‘privileged structure'. Mini Rev. Med. Chem. 9, 782–793. 10.2174/138955709788452649 [DOI] [PubMed] [Google Scholar]
- Di Pietro A., Conseil G., Perez-Victoria J. M., Dayan G., Baubichon-Cortay H., Trompier D., et al. (2002). Modulation by flavonoids of cell multidrug resistance mediated by P-glycoprotein and related ABC transporters. Cell. Mol. Life Sci. 59, 307–322. 10.1007/s00018-002-8424-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dong Q. H., Zheng S., Xu R. Z., Lu Q., He L. (2004). Study on effect of berbamine on multidrug resistance leukemia K562/Adr cells. Chin. J. Integrated Tradit. West. Med. 24, 820–822. [PubMed] [Google Scholar]
- Doyle L. A., Yang W., Abruzzo L. V., Krogmann T., Gao Y., Rishi A. K., et al. (1998). A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc. Natl. Acad. Sci. 95, 15665–15670. 10.1073/pnas.95.26.15665 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duarte N., Gyémánt N., Abreu P. M., Molnár J., Ferreira M. J. U. (2006). New macrocyclic lathyrane diterpenes, from Euphorbia lagascae, as inhibitors of multidrug resistance of tumour cells. Planta Med. 72, 162–168. 10.1055/s-2005-873196 [DOI] [PubMed] [Google Scholar]
- Duarte N., Járdánházy A., Molnár J., Hilgeroth A., Ferreira M. J. U. (2008). Synergistic interaction between p-glycoprotein modulators and epirubicine on resistant cancer cells. Bioorg. Med. Chem. 16, 9323–9330. 10.1016/j.bmc.2008.08.071 [DOI] [PubMed] [Google Scholar]
- Durmus S., Hendrikx J. J., Schinkel A. H. (2015). “Apical ABC transporters and cancer chemotherapeutic drug disposition,” in Advances in Cancer Research (San Diego, USA: Academic Press; ), 1–41. [DOI] [PubMed] [Google Scholar]
- Efferth T., Davey M., Olbrich A., Rücker G., Gebhart E., Davey R. (2002). Activity of drugs from traditional Chinese medicine toward sensitive and MDR1-or MRP1-overexpressing multidrug-resistant human CCRF-CEM leukemia cells. Blood Cells Mol. Dis. 28, 160–168. 10.1006/bcmd.2002.0492 [DOI] [PubMed] [Google Scholar]
- Eid S. Y., El-Readi M. Z., Wink M. (2012). Carotenoids reverse multidrug resistance in cancer cells by interfering with ABC-transporters. Phytomedicine 19, 977–987. 10.1016/J.PHYMED.2012.05.010 [DOI] [PubMed] [Google Scholar]
- Eid S. Y., El-Readi M. Z., Eldin E. E. M. N., Fatani S. H., Wink M. (2013). Influence of combinations of digitonin with selected phenolics, terpenoids, and alkaloids on the expression and activity of P-glycoprotein in leukaemia and colon cancer cells. Phytomedicine 21, 47–61. 10.1016/J.PHYMED.2013.07.019 [DOI] [PubMed] [Google Scholar]
- El-Readi M. Z., Hamdan D., Farrag N., El-Shazly A., Wink M. (2010). Inhibition of P-glycoprotein activity by limonin and other secondary metabolites from Citrus species in human colon and leukaemia cell lines. Eur. J. Pharmacol. 626, 139–145. 10.1016/j.ejphar.2009.09.040 [DOI] [PubMed] [Google Scholar]
- El-Readi M. Z., Eid S., Abdelghany A. A., Al-Amoudi H. S., Efferth T., Wink M. (2019). Resveratrol mediated cancer cell apoptosis, and modulation of multidrug resistance proteins and metabolic enzymes. Phytomedicine 55, 269–281. 10.1016/J.PHYMED.2018.06.046 [DOI] [PubMed] [Google Scholar]
- Fantini M., Benvenuto M., Masuelli L., Frajese G. V., Tresoldi I., Modesti A., et al. (2015). In vitro and in vivo antitumoral effects of combinations of polyphenols, or polyphenols and anticancer drugs: Perspectives on cancer treatment. Int. J. Mol. Sci. 16, 9236–9282. 10.3390/ijms16059236 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Farabegoli F., Papi A., Bartolini G., Ostan R., Orlandi M. (2010). (-)-Epigallocatechin-3-gallate downregulates P-gp and BCRP in a tamoxifen resistant MCF-7 cell line. Phytomedicine 17, 356–362. 10.1016/j.phymed.2010.01.001 [DOI] [PubMed] [Google Scholar]
- Ferreira R. J., Baptista R., Moreno A., Madeira P. G., Khonkarn R., Baubichon-Cortay H., et al. (2018). Optimizing the flavanone core toward new selective nitrogen-containing modulators of ABC transporters. Future Med. Chem. 10, 725–741. 10.4155/fmc-2017-0228 [DOI] [PubMed] [Google Scholar]
- Fong W. F., Wan C. K., Zhu G. Y., Chattopadhyay A., Dey S., Zhao Z., et al. (2007). Schisandrol A from Schisandra chinensis reverses P-glycoprotein-mediated multidrug resistance by affecting Pgp-substrate complexes. Planta Med. 73, 212–220. 10.1055/s-2007-967120 [DOI] [PubMed] [Google Scholar]
- Gade D. R., Makkapati A., Yarlagadda R. B., Peters G. J., Sastry B. S., Prasad V. R. (2018). Elucidation of chemosensitization effect of acridones in cancer cell lines: Combined pharmacophore modeling, 3D QSAR, and molecular dynamics studies. Comput. Biol. Chem. 74, 63–75. 10.1016/j.compbiolchem.2018.02.014 [DOI] [PubMed] [Google Scholar]
- Gan C. Y., Low Y. Y., Etoh T., Hayashi M., Komiyama K., Kam T. S. (2009). Leuconicines A− G and (−)-Eburnamaline, Biologically Active Strychnan and Eburnan Alkaloids from Leuconotis. J. Nat. Prod. 72, 2098–2103. 10.1021/np900576b [DOI] [PubMed] [Google Scholar]
- Gan C. Y., Yoganathan K., Sim K. S., Low Y. Y., Lim S. H., Kam T. S. (2014). Corynanthean, eburnan, secoleuconoxine, and pauciflorine alkaloids from Kopsia pauciflora. Phytochemistry 108, 234–242. 10.1016/j.phytochem.2014.09.014 [DOI] [PubMed] [Google Scholar]
- Ganta S., Amiji M. (2009). Coadministration of paclitaxel and curcumin in nanoemulsion formulations to overcome multidrug resistance in tumor cells. Mol. Pharm. 6, 928–939. 10.1021/mp800240j [DOI] [PubMed] [Google Scholar]
- Gao N., Zhang Y., Mao J. Q., Li G. Q., Zhou W., Gao B., et al. (2008). Effects of some traditional Chinese drugs on Mdr1 gene and its expression product in K562/A02 cells. J. Exp. Hematol. 16, 785–789. [PubMed] [Google Scholar]
- Gawali V. S., Simeonov S., Drescher M., Knott T., Scheel O., Kudolo J., et al. (2017). C2-modified sparteine derivatives are a new class of potentially long-acting sodium channel blockers. ChemMedChem 12, 1819–1822. 10.1002/cmdc.201700568 [DOI] [PubMed] [Google Scholar]
- Gilbert K. (2001). Review of Plant Secondary Metabolism, by David S. Seigler. Plant Growth Regul. 34, 149. 10.1023/A:1013354907356 [DOI] [Google Scholar]
- Guo Q., Nan X. X., Yang J. R., Yi L., Liang B. L., Wei Y. B., et al. (2013). Triptolide inhibits the multidrug resistance in prostate cancer cells via the downregulation of MDR1 expression. Neoplasma 60, 598–604. 10.4149/neo_2013_077 [DOI] [PubMed] [Google Scholar]
- Gyémánt N., Tanaka M., Antus S., Hohmann J., Csuka O., Mándoky L., et al. (2005). and In vitro search for synergy between flavonoids and epirubicin on multidrug-resistant cancer cells. Vivo 19, 367–374. [PubMed] [Google Scholar]
- Gyémánt N., Tanaka M., Molnár P., Deli J., Mándoky L., Molnár J. (2006). Reversal of multidrug resistance of cancer cells in vitro: modification of drug resistance by selected carotenoids. Anticancer Res. 26, 367–374. [PubMed] [Google Scholar]
- Hänsel R., Sticher O. (Eds.) (2009). Pharmakognosie-Phytopharmazie (Heidelberg, Germany: Springer-Verlag; ). [Google Scholar]
- Han Y., Tan T. M. C., Lim L. Y. (2006). Effects of capsaicin on P-gp function and expression in Caco-2 cells. Biochem. Pharmacol. 71, 1727–1734. 10.1016/j.bcp.2006.03.024 [DOI] [PubMed] [Google Scholar]
- He L., Zhao C., Yan M., Zhang L. Y., Xia Y. Z. (2009). Inhibition of P-glycoprotein function by procyanidine on blood–brain barrier. Phytother. Res. 23, 933–937. 10.1002/ptr.2781 [DOI] [PubMed] [Google Scholar]
- He L., Yang J., Hu L. (2010). Transmembrane transport activity of paclitaxel regulated by fangchinoline in MDR1-mDCK II cells. China J. Chin. Materia Med. 35, 1478–1481. [PubMed] [Google Scholar]
- Hegde R., Thimmaiah P., Yerigeri M. C., Krishnegowda G., Thimmaiah K. N., Houghton P. J. (2004). Anti-calmodulin acridone derivatives modulate vinblastine resistance in multidrug resistant (MDR) cancer cells. Eur. J. Med. Chem. 39, 161–177. 10.1016/j.ejmech.2003.12.001 [DOI] [PubMed] [Google Scholar]
- Hsiao S. H., Lu Y. J., Yang C. C., Tuo W. C., Li Y. Q., Huang Y. H., et al. (2016). Hernandezine, a bisbenzylisoquinoline alkaloid with selective inhibitory activity against multidrug-resistance-linked ATP-binding cassette drug transporter ABCB1. J. Nat. Prod. 79, 2135–2142. 10.1021/acs.jnatprod.6b00597 [DOI] [PubMed] [Google Scholar]
- Hu Y. J., Shen X. L., Lu H. L., Zhang Y. H., Huang X. A., Fu L. C., et al. (2008). Tenacigenin B derivatives reverse P-glycoprotein-mediated multidrug resistance in HepG2/Dox cells. J. Nat. Prod. 71, 1049–1051. 10.1021/np070458f [DOI] [PubMed] [Google Scholar]
- Huang M., Jin J., Sun H., Liu G. T. (2008). Reversal of P-glycoprotein-mediated multidrug resistance of cancer cells by five schizandrins isolated from the Chinese herb Fructus schizandrae. Canc. Chemother. Pharmacol. 62, 1015–1026. 10.1007/s00280-008-0691-0 [DOI] [PubMed] [Google Scholar]
- Huang F., Wu X. N., Chen J. I. E., Wang W. X., Lu Z. F. (2014). Resveratrol reverses multidrug resistance in human breast cancer doxorubicin-resistant cells. Exp. Ther. Med. 7, 1611–1616. 10.3892/etm.2014.1662 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ikegawa T., Ushigome F., Koyabu N., Morimoto S., Shoyama Y., Naito M., et al. (2000). Inhibition of P-glycoprotein by orange juice components, polymethoxyflavones in adriamycin-resistant human myelogenous leukemia (K562/ADM) cells. Canc. Lett. 160, 21–28. 10.1016/S0304-3835(00)00549-8 [DOI] [PubMed] [Google Scholar]
- Imai Y., Tsukahara S., Asada S., Sugimoto Y. (2004). Phytoestrogens/flavonoids reverse breast cancer resistance protein/ABCG2-mediated multidrug resistance. Canc. Res. 64, 4346–4352. 10.1158/0008-5472.CAN-04-0078 [DOI] [PubMed] [Google Scholar]
- Incardona J. P., Gaffield W., Kapur R. P., Roelink H. (1998). The teratogenic Veratrum alkaloid cyclopamine inhibits sonic hedgehog signal transduction. Development 125, 3553–3562. [DOI] [PubMed] [Google Scholar]
- Ivanova A., Serly J., Dinchev D., Ocsovszki I., Kostova I., Molnár J. (2009). Screening of some saponins and phenolic components of Tribulus terrestris and Smilax excelsa as MDR modulators. Vivo 23, 545–550. [PubMed] [Google Scholar]
- Ivanova A., Serly J., Christov V., Stamboliyska B., Molnaár J. (2011). Alkaloids derived from genus Veratrum and Peganum of Mongolian origin as multidrug resistance inhibitors of cancer cells. Fitoterapia 82, 570–575. 10.1016/J.FITOTE.2011.01.015 [DOI] [PubMed] [Google Scholar]
- Jones P. M., George A. M. (2002). Mechanism of ABC transporters: a molecular dynamics simulation of a well characterized nucleotide-binding subunit. Proc. Natl. Acad. Sci. 99, 12639–12644. 10.1073/pnas.152439599 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kam T. S., Subramaniam G., Sim K. M., Yoganathan K., Koyano T., Toyoshima M., et al. (1998). Reversal of multidrug resistance (MDR) by aspidofractinine-type indole alkaloids. Bioorg. Med. Chem. Lett. 8, 2769–2772. 10.1016/S0960-894X(98)00486-7 [DOI] [PubMed] [Google Scholar]
- Kamath K., Wilson L., Cabral F., Jordan M. A. (2005). βIII-tubulin induces paclitaxel resistance in association with reduced effects on microtubule dynamic instability. J. Biol. Chem. 280, 12902–12907. 10.1074/jbc.M414477200 [DOI] [PubMed] [Google Scholar]
- Kasaian J., Mosaffa F., Behravan J., Masullo M., Piacente S., Ghandadi M., et al. (2015). Reversal of P-glycoprotein-mediated multidrug resistance in MCF-7/Adr cancer cells by sesquiterpene coumarins. Fitoterapia 103, 149–154. 10.1016/j.fitote.2015.03.025 [DOI] [PubMed] [Google Scholar]
- Kashiwada Y., Nishimura K., Kurimoto S., Takaishi Y. (2011). New 29-nor-cycloartanes with a 3,4-seco- and a novel 2,3-seco-structure from the leaves of Sinocalycanthus chinensis. Bioorg. Med. Chem. 19, 2790–2796. 10.1016/j.bmc.2011.03.055 [DOI] [PubMed] [Google Scholar]
- Kavallaris M., Tait A. S., Walsh B. J., He L., Horwitz S. B., Norris M. D., et al. (2001). Multiple microtubule alterations are associated with Vinca alkaloid resistance in human leukemia cells. Canc. Res. 61, 5803–5809. [PubMed] [Google Scholar]
- Kelly R. J., Draper D., Chen C. C., Robey R. W., Figg W. D., Piekarz R. L., et al. (2011). A pharmacodynamic study of docetaxel in combination with the P-glycoprotein antagonist tariquidar (XR9576) in patients with lung, ovarian, and cervical cancer. Clin. Canc. Res. 17, 569–580. 10.1158/1078-0432.CCR-10-1725 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim S. W., Kwon H. Y., Chi D. W., Shim J. H., Park J. D., Lee Y. H., et al. (2003). Reversal of P-glycoprotein-mediated multidrug resistance by ginsenoside Rg3. Biochem. Pharmacol. 65, 75–82. 10.1016/S0006-2952(02)01446-6 [DOI] [PubMed] [Google Scholar]
- Kim E.-H., Min H.-Y., Chung H.-J., Song J., Park H.-J., Kim S., et al. (2012). Anti-proliferative activity and suppression of P-glycoprotein by (–)-antofine, a natural phenanthroindolizidine alkaloid, in paclitaxel-resistant human lung cancer cells. Food Chem. Toxicol. 50, 1060–1065. 10.1016/J.FCT.2011.11.008 [DOI] [PubMed] [Google Scholar]
- Kitagawa S., Nabekura T., Kamiyama S. (2004). Inhibition of P-glycoprotein function by tea catechins in KB-C2 cells. J. Pharm. Pharmacol. 56, 1001–1005. 10.1211/0022357044003 [DOI] [PubMed] [Google Scholar]
- Kitagawa S., Nabekura T., Takahashi T., Nakamura Y., Sakamoto H., Tano H., et al. (2005). Structure–activity relationships of the inhibitory effects of flavonoids on P-glycoprotein-mediated transport in KB-C2 cells. Biol. Pharm. Bull. 28, 2274–2278. 10.1248/bpb.28.2274 [DOI] [PubMed] [Google Scholar]
- Kitagawa S., Takahashi T., Nabekura T., Tachikawa E., Hasegawa H. (2007). Inhibitory effects of ginsenosides and their hydrolyzed metabolites on daunorubicin transport in KB-C2 cells. Biol. Pharm. Bull. 30, 1979–1981. 10.1248/bpb.30.1979 [DOI] [PubMed] [Google Scholar]
- Koes R. E., Quattrocchio F., Mol J. N. (1994). The flavonoid biosynthetic pathway in plants: function and evolution. BioEssays 16, 123–132. 10.1002/bies.950160209 [DOI] [Google Scholar]
- König J., Hartel M., Nies A. T., Martignoni M. E., Guo J., Büchler M. W., et al. (2005). Expression and localization of human multidrug resistance protein (ABCC) family members in pancreatic carcinoma. Int. J. Canc. 115, 359–367. 10.1002/ijc.20831 [DOI] [PubMed] [Google Scholar]
- Körper S., Wink M., Fink R. H. (1998). Differential effects of alkaloids on sodium currents of isolated single skeletal muscle fibers. FEBS Lett. 436, 251–255. 10.1016/S0014-5793(98)01135-1 [DOI] [PubMed] [Google Scholar]
- Kukula-Koch W. A., Widelski J. (2017). “Alkaloids,” in Pharmacognosy (San Diego, USA: Academic Press; ), 163–198. [Google Scholar]
- Kurimoto S. I., Kashiwada Y., Lee K. H., Takaishi Y. (2011. a). Triterpenes and a triterpene glucoside from Dysoxylum cumingianum. Phytochemistry 72, 2205–2211. 10.1016/j.phytochem.2011.08.002 [DOI] [PubMed] [Google Scholar]
- Kurimoto S. I., Kashiwada Y., Morris-Natschke S. L., Lee K. H., Takaishi Y. (2011. b). Dyscusins A—C, three new steroids from the leaves of Dysoxylum cumingianum. Chem. Pharm. Bull. 59, 1303–1306. 10.1248/cpb.59.1303 [DOI] [PubMed] [Google Scholar]
- Lei Y., Tan J., Wink M., Ma Y., Li N., Su G. (2013). An isoquinoline alkaloid from the Chinese herbal plant Corydalis yanhusuo W.T. Wang inhibits P-glycoprotein and multidrug resistance-associate protein 1. Food Chem. 136, 1117–1121. 10.1016/j.foodchem.2012.09.059 [DOI] [PubMed] [Google Scholar]
- Lemmer B., Brune K. (2004). Pharmakotherapie (Heidelberg, Germany: Springer-Verlag; ). [Google Scholar]
- Li L., Stanton J. D., Tolson A. H., Luo Y., Wang H. (2009). Bioactive terpenoids and flavonoids from Ginkgo biloba extract induce the expression of hepatic drug-metabolizing enzymes through pregnane X receptor, constitutive androstane receptor, and aryl hydrocarbon receptor-mediated pathways. Pharmaceut. Res. 26, 872. 10.1007/s11095-008-9788-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y., Wang Q., Yao X., Li Y. (2010). Induction of CYP3A4 and MDR1 gene expression by baicalin, baicalein, chlorogenic acid, and ginsenoside Rf through constitutive androstane receptor-and pregnane X receptor-mediated pathways. Eur. J. Pharmacol. 640, 46–54. 10.1016/j.ejphar.2010.05.017 [DOI] [PubMed] [Google Scholar]
- Li S., Lei Y., Jia Y., Li N., Wink M., Ma Y. (2011). Piperine, a piperidine alkaloid from Piper nigrum re-sensitizes P-gp, MRP1 and BCRP dependent multidrug resistant cancer cells. Phytomedicine 19, 83–87. 10.1016/j.phymed.2011.06.031 [DOI] [PubMed] [Google Scholar]
- Li H., Krstin S., Wang S., Wink M. (2018. a). Capsaicin and piperine can overcome multidrug resistance in cancer cells to doxorubicin. Molecules 23, 557. 10.3390/molecules23030557 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li H., Krstin S., Wink M. (2018. b). Modulation of multidrug resistant in cancer cells by EGCG, tannic acid and curcumin. Phytomedicine 50, 213–222. 10.1016/j.phymed.2018.09.169 [DOI] [PubMed] [Google Scholar]
- Liang G., Tang A., Lin X., Li L., Zhang S., Huang Z., et al. (2010). Green tea catechins augment the antitumor activity of doxorubicin in an in vivo mouse model for chemoresistant liver cancer. Int. J. Oncol. 37, 111–123. 10.3892/ijo_00000659 [DOI] [PubMed] [Google Scholar]
- Liao D., Zhang W., Gupta P., Lei Z. N., Wang J. Q., Cai C. Y., et al. (2019). Tetrandrine Interaction with ABCB1 Reverses Multidrug Resistance in Cancer Cells Through Competition with Anti-Cancer Drugs Followed by Downregulation of ABCB1 Expression. Molecules 24, 4383. 10.3390/molecules24234383 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Limtrakul P., Anuchapreeda S., Buddhasukh D. (2004). Modulation of human multidrug-resistance MDR-1 gene by natural curcuminoids. BMC Canc. 4, 13. 10.1186/1471-2407-4-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Limtrakul P., Chearwae W., Shukla S., Phisalphong C., Ambudkar S. V. (2007). Modulation of function of three ABC drug transporters, P-glycoprotein (ABCB1), mitoxantrone resistance protein (ABCG2) and multidrug resistance protein 1 (ABCC1) by tetrahydrocurcumin, a major metabolite of curcumin. Mol. Cell. Biochem. 296, 85–95. 10.1007/s11010-006-9302-8 [DOI] [PubMed] [Google Scholar]
- Lin H. L., Liu T. Y., Lui W. Y., Chi C. W. (1999. a). Up-regulation of multidrug resistance transporter expression by berberine in human and murine hepatoma cells. Canc. Interdiscipl. Int. J. Am. Canc. Soc 85, 1937–1942. [DOI] [PubMed] [Google Scholar]
- Lin H.-L., Liu T.-Y., Wu C.-W., Chi C.-W. (1999. b). Berberine modulates expression of MDR1 gene product and the responses of digestive track cancer cells to paclitaxel. Br. J. Canc. 81, 416–422. 10.1038/sj.bjc.6690710 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu C. M., Kao C. L., Tseng Y. T., Lo Y. C., Chen C. Y. (2017). Ginger phytochemicals inhibit cell growth and modulate drug resistance factors in docetaxel resistant prostate cancer cell. Molecules 22, 1477. 10.3390/molecules22091477 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu J. J., Dang Y. Y., Huang M., Xu W. S., Chen X. P., Wang Y. T. (2012). Anti-cancer properties of terpenoids isolated from Rhizoma curcumae – a review. J. Ethnopharmacol. 143, 406–411. 10.1016/j.jep.2012.07.009 [DOI] [PubMed] [Google Scholar]
- Lu W. D., Qin Y., Yang C., Li L. (2013). Effect of curcumin on human colon cancer multidrug resistance in vitro and in vivo. Clinics 68, 694–701. 10.6061/clinics/2013(05)18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma Y., Wink M. (2008). Lobeline, a piperidine alkaloid from Lobelia can reverse P-gp dependent multidrug resistance in tumor cells. Phytomedicine 15, 754–758. 10.1016/j.phymed.2007.11.028 [DOI] [PubMed] [Google Scholar]
- Ma Y., Wink M. (2010). The beta-carboline alkaloid harmine inhibits BCRP and can reverse resistance to the anticancer drugs mitoxantrone and camptothecin in breast cancer cells. Phytother. Res. 24, 146–149. 10.1002/ptr.2860 [DOI] [PubMed] [Google Scholar]
- Ma W., Feng S., Yao X., Yuan Z., Liu L., Xie Y. (2015). Nobiletin enhances the efficacy of chemotherapeutic agents in ABCB1 overexpression cancer cells. Sci. Rep. 5, 18789. 10.1038/srep18789 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martins A., Vasas A., Schelz Z., Viveiros M., Molnár J., Hohmann J., et al. (2010). Constituents of Carpobrotus edulis inhibit P-glycoprotein of MDR1-transfected mouse lymphoma cells. Anticancer Res. 30, 829–835. [PubMed] [Google Scholar]
- Mayur Y. C., Padma T., Parimala B. H., Chandramouli K. H., Jagadeesh S., Gowda N. M., et al. (2006). Sensitization of multidrug resistant (MDR) cancer cells to vinblastine by novel acridones: correlation between anti-calmodulin activity and anti-MDR activity. Med. Chem. 2, 63–77. 10.2174/157340606775197732 [DOI] [PubMed] [Google Scholar]
- Mayur Y. C. (2015). “Designing of drug molecules for reversing P-glycoprotein (Pgp) mediated drug resistance in cancer cells,” in Frontiers in Anti-Cancer Drug Discovery. Eds. Ata-ur- R., Chaudhary M. I. (Sharjah, UAE: Bentham Science Publishers; ), 157–198. [Google Scholar]
- Mi Q., Cui B., Silva G. L., Lantvit D., Lim E., Chai H., et al. (2001). Pervilleine A, a novel tropane alkaloid that reverses the multidrug-resistance phenotype. Canc. Res. 61, 4030–4037. [PubMed] [Google Scholar]
- Mi Q., Cui B., Silva G. L., Lantvit D., Lim E., Chai H., et al. (2002). Pervilleines B and C, new tropane alkaloid aromatic esters that reverse the multidrug-resistance in the hollow fiber assay. Canc. Lett. 184, 13–20. 10.1016/S0304-3835(02)00202-1 [DOI] [PubMed] [Google Scholar]
- Min Y. D., Choi S. U., Lee K. R. (2006). Aporphine alkaloids and their reversal activity of multidrug resistance (MDR) from the stems and rhizomes of Sinomenium acutum. Arch. Pharm. Res. 29, 627–632. 10.1007/BF02968246 [DOI] [PubMed] [Google Scholar]
- Min Y. D., Kwon H. C., Yang M. C., Lee K. H., Choi S. U., Lee K. R. (2007). Isolation of limonoids and alkaloids from Phellodendron amurense and their multidrug resistance (MDR) reversal activity. Arch. Pharmacal Res. 30, 58–63. 10.1007/BF02977779 [DOI] [PubMed] [Google Scholar]
- Mitani Y., Satake K., Tsukamoto M., Nakamura I., Kadioglu O., Teruya T., et al. (2018). Epimagnolin A, a tetrahydrofurofuranoid lignan from Magnolia fargesii, reverses ABCB1-mediated drug resistance. Phytomedicine 51, 112–119. 10.1016/j.phymed.2018.06.030 [DOI] [PubMed] [Google Scholar]
- Mohanlal R., Sun Y., Kloecker G., Feinstein T., Shi Y., Han B., et al. (2019). P2. 01-23 DUBLIN-3, a phase (Ph) III trial comparing the plinabulin (P)/docetaxel (D) combination with D alone in stage IIIb/IV NSCLC. J. Thorac. Oncol. 14, S647–S648. 10.1016/j.jtho.2019.08.1367 [DOI] [Google Scholar]
- Molnár J., Szabo D., Pusztai R., Mucsi I., Berek L., Ocsovszki I., et al. (2000). Membrane associated antitumor effects of crocine-, ginsenoside-and cannabinoid derivates. Anticancer Res. 20, 861–867. [PubMed] [Google Scholar]
- Molnár J., Gyémánt N., Tanaka M., Hohmann J., Bergmann-Leitner E., Molnár P., et al. (2006). Inhibition of multidrug resistance of cancer cells by natural diterpenes, triterpenes and carotenoids. Curr. Pharmaceut. Des. 12, 287–311. 10.2174/138161206775201893 [DOI] [PubMed] [Google Scholar]
- Molnár J., Engi H., Hohmann J., Molnár P., Deli J., Wesolowska O., et al. (2010). Reversal of multidrug resistance by natural substances from plants. Curr. Top. Med. Chem. 10, 1757–1768. 10.2174/156802610792928103 [DOI] [PubMed] [Google Scholar]
- Moussavi M., Haddad F., Rassouli F. B., Iranshahi M., Soleymanifard S. (2017). Synergy between auraptene, ionizing radiation, and anticancer drugs in colon adenocarcinoma cells. Phytother. Res. 31, 1369–1375. 10.1002/ptr.5863 [DOI] [PubMed] [Google Scholar]
- Murahari M., Kharkar P. S., Lonikar N., Mayur Y. C. (2017). Design, synthesis, biological evaluation, molecular docking and QSAR studies of 2, 4-dimethylacridones as anticancer agents. Eur. J. Med. Chem. 130, 154–170. 10.1016/j.ejmech.2017.02.022 [DOI] [PubMed] [Google Scholar]
- Nabekura T., Kamiyama S., Kitagawa S. (2005). Effects of dietary chemopreventive phytochemicals on P-glycoprotein function. Biochem. Biophys. Res. Commun. 327, 866–870. 10.1016/j.bbrc.2004.12.081 [DOI] [PubMed] [Google Scholar]
- Nabekura T., Yamaki T., Ueno K., Kitagawa S. (2008. a). Inhibition of P-glycoprotein and multidrug resistance protein 1 by dietary phytochemicals. Canc. Chemother. Pharmacol. 62, 867–873. 10.1007/s00280-007-0676-4 [DOI] [PubMed] [Google Scholar]
- Nabekura T., Yamaki T., Kitagawa S. (2008. b). Effects of chemopreventive citrus phytochemicals on human P-glycoprotein and multidrug resistance protein 1. Eur. J. Pharmacol. 600, 45–49. 10.1016/j.ejphar.2008.10.025 [DOI] [PubMed] [Google Scholar]
- Nabekura T., Yamaki T., Ueno K., Kitagawa S. (2008. c). Effects of plant sterols on human multidrug transporters ABCB1 and ABCC1. Biochem. Biophys. Res. Commun. 369, 363–368. 10.1016/j.bbrc.2008.02.026 [DOI] [PubMed] [Google Scholar]
- Nabekura T., Yamaki T., Hiroi T., Ueno K., Kitagawa S. (2010). Inhibition of anticancer drug efflux transporter P-glycoprotein by rosemary phytochemicals. Pharmacol. Res. 61, 259–263. 10.1016/j.phrs.2009.11.010 [DOI] [PubMed] [Google Scholar]
- Najar I. A., Sachin B. S., Sharma S. C., Satti N. K., Suri K. A., Johri R. K. (2010). Modulation of P-glycoprotein ATPase activity by some phytoconstituents. Phytother. Res. 24, 454–458. 10.1002/ptr.2951 [DOI] [PubMed] [Google Scholar]
- Namanja H. A., Emmert D., Pires M. M., Hrycyna C. A., Chmielewski J. (2009). Inhibition of human P-glycoprotein transport and substrate binding using a galantamine dimer. Biochem. Biophys. Res. Commun. 388, 672–676. 10.1016/J.BBRC.2009.08.056 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neto S., Duarte N., Pedro C., Spengler G., Molnár J., Ferreira M. J. U. (2019). Effective MDR reversers through phytochemical study of Euphorbia boetica. Phytochem. Anal. 30, 498–511. 10.1002/pca.2841 [DOI] [PubMed] [Google Scholar]
- Nguyen H., Zhang S., Morris M. E. (2003). Effect of flavonoids on MRP1-mediated transport in Panc-1 cells. J. Pharmaceut. Sci. 92, 250–257. 10.1002/jps.10283 [DOI] [PubMed] [Google Scholar]
- Nieri P., Romiti N., Adinolfi B., Chicca A., Massarelli I., Chieli E. (2006). Modulation of P-glycoprotein activity by cannabinoid molecules in HK-2 renal cells. Br. J. Pharmacol. 148, 682. 10.1038/sj.bjp.0706778 [DOI] [PMC free article] [PubMed] [Google Scholar]
- O'Connor P. M., Jackman J., Bae I., Myers T. G., Fan S., Mutoh M., et al. (1997). Characterization of the p53 tumor suppressor pathway in cell lines of the National Cancer Institute anticancer drug screen and correlations with the growth-inhibitory potency of 123 anticancer agents. Canc. Res. 57, 4285–4300. [PubMed] [Google Scholar]
- Ohtani H., Ikegawa T., Honda Y., Kohyama N., Morimoto S., Shoyama Y., et al. (2007). Effects of various methoxyflavones on vincristine uptake and multidrug resistance to vincristine in P-gp-overexpressing K562/ADM cells. Pharmaceut. Res. 24, 1936–1943. 10.1007/s11095-007-9320-6 [DOI] [PubMed] [Google Scholar]
- Osman A. M. G., Chittiboyina A. G., Khan I. A. (2013). “Foodborne infections and intoxications: Chapter 32,” in Plant Toxins (San Diego, USA: Academic Press; ). [Google Scholar]
- Panche A. N., Diwan A. D., Chandra S. R. (2016). Flavonoids: an overview. J. Nutr. Sci. 5 (E47), 1–15. 10.1017/jns.2016.41 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pearce H. L., Safa A. R., Bach N. J., Winter M. A., Cirtain M. C., Beck W. T. (1989). Essential features of the P-glycoprotein pharmacophore as defined by a series of reserpine analogs that modulate multidrug resistance. Proc. Natl. Acad. Sci. U.S.A. 86, 5128–5132. 10.1073/PNAS.86.13.5128 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pires M. M., Emmert D., Hrycyna C. A., Chmielewski J. (2009). Inhibition of P-glycoprotein-mediated paclitaxel resistance by reversibly linked quinine homodimers. Mol. Pharmacol. 75, 92–100. 10.1124/mol.108.050492 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reis M. A., Ahmed O. B., Spengler G., Molnár J., Lage H., Ferreira M. J. U. (2016). Jatrophane diterpenes and cancer multidrug resistance–ABCB1 efflux modulation and selective cell death induction. Phytomedicine 23, 968–978. 10.1016/j.phymed.2016.05.007 [DOI] [PubMed] [Google Scholar]
- Rethy B., Hohmann J., Minorics R., Varga A., Ocsovszki I., Molnár J., et al. (2008). Antitumour properties of acridone alkaloids on a murine lymphoma cell line. Anticancer Res. 28, 2737–2743. [PubMed] [Google Scholar]
- Reyes-Esparza J., Morales A. I. G., González-Maya L., Rodríguez-Fragoso L. (2015). (-)-Epigallocatechin-3-gallate modulates the activity and expression of P-glycoprotein in breast cancer cells. J. Pharmacol. Clin. Toxicol. 3, 1044. [Google Scholar]
- Roberts M. F., Wink M. (1998). Alkaloids: Biochemistry, Ecology, and Medicinal Applications (New York, USA: Plenum Press; ). [Google Scholar]
- Ruenitz P. C., Mokler C. M. (1977). Analogs of sparteine. 5. antiarrhythmic activity of selected N, N'-disubstituted bispidines. J. Med. Chem. 20, 1668–1671. 10.1021/jm00222a026 [DOI] [PubMed] [Google Scholar]
- Saeed M., Kadioglu O., Khalid H., Sugimoto Y., Efferth T. (2015). Activity of the dietary flavonoid, apigenin, against multidrug-resistant tumor cells as determined by pharmacogenomics and molecular docking. J. Nutr. Biochem. 26, 44–56. 10.1016/j.jnutbio.2014.09.008 [DOI] [PubMed] [Google Scholar]
- Sampson K. E., Wolf C. L., Abraham I. (1993). Staurosporine reduces P-glycoprotein expression and modulates multidrug resistance. Canc. Lett. 68, 7–14. 10.1016/0304-3835(93)90213-S [DOI] [PubMed] [Google Scholar]
- Sergent T., Garsou S., Schaut A., De Saeger S., Pussemier L., Van Peteghem C., et al. (2005). Differential modulation of ochratoxin A absorption across Caco-2 cells by dietary polyphenols, used at realistic intestinal concentrations. Toxicol. Lett. 159, 60–70. 10.1016/j.toxlet.2005.04.013 [DOI] [PubMed] [Google Scholar]
- Shen X., Chen G., Zhu G., Fong W. F. (2006). (±)-3′-O, 4′-O-dicynnamoyl-cis-khellactone, a derivative of (±)-praeruptorin A, reverses P-glycoprotein mediated multidrug resistance in cancer cells. Bioorg. Med. Chem. 14, 7138–7145. 10.1016/j.bmc.2006.06.066 [DOI] [PubMed] [Google Scholar]
- Shen H., Xu W., Chen Q., Wu Z., Tang H., Wang F. (2010). Tetrandrine prevents acquired drug resistance of K562 cells through inhibition of MDR1 gene transcription. J. Canc. Res. Clin. Oncol. 136, 659–665. 10.1007/s00432-009-0704-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sheu M. T., Liou Y. B., Kao Y. H., Lin Y. K., Ho H. O. (2010). A quantitative structure–activity relationship for the modulation effects of flavonoids on P-glycoprotein-mediated transport. Chem. Pharmaceut. Bull. 58, 1187–1194. 10.1248/cpb.58.1187 [DOI] [PubMed] [Google Scholar]
- Simstein R., Burow M., Parker A., Weldon C., Beckman B. (2003). Apoptosis, chemoresistance, and breast cancer: insights from the MCF-7 cell model system. Exp. Biol. Med. 228, 995–1003. 10.1177/153537020322800903 [DOI] [PubMed] [Google Scholar]
- Slaninová I., Březinová L., Koubíková L., Slanina J. (2009). Dibenzocyclooctadiene lignans overcome drug resistance in lung cancer cells–study of structure–activity relationship. Toxicol. Vitro 23, 1047–1054. 10.1016/j.tiv.2009.06.008 [DOI] [PubMed] [Google Scholar]
- Smith E. W., Maibach H. I. (1995). Percutaneous Penetration Enhancers (Boca Raton, USA: CRC Press; ). [Google Scholar]
- Smith E. C., Kaatz G. W., Seo S. M., Wareham N., Williamson E. M., Gibbons S. (2007). The phenolic diterpene totarol inhibits multidrug efflux pump activity in Staphylococcus aureus. Antimicrob. Agents Chemother. 51, 4480–4483. 10.1128/AAC.00216-07 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stermitz F. R., Scriven L. N., Tegos G., Lewis K. (2002). Two flavonols from Artemisa annua which potentiate the activity of berberine and norfloxacin against a resistant strain of Staphylococcus aureus. Planta Med. 68, 1140–1141. 10.1055/s-2002-36347 [DOI] [PubMed] [Google Scholar]
- Su S., Cheng X., Wink M. (2015). Natural lignans from Arctium lappa modulate P-glycoprotein efflux function in multidrug resistant cancer cells. Phytomedicine 22, 301–307. 10.1016/J.PHYMED.2014.12.009 [DOI] [PubMed] [Google Scholar]
- Sun Y. F., Wink M. (2014). Tetrandrine and fangchinoline, bisbenzylisoquinoline alkaloids from Stephania tetrandra can reverse multidrug resistance by inhibiting P-glycoprotein activity in multidrug resistant human cancer cells. Phytomedicine 21, 1110–1119. 10.1016/J.PHYMED.2014.04.029 [DOI] [PubMed] [Google Scholar]
- Sun H. X., Xie Y., Ye Y. P. (2009). Advances in saponin-based adjuvants. Vaccine 27, 1787–1796. 10.1016/j.vaccine.2009.01.091 [DOI] [PubMed] [Google Scholar]
- Sun J., Yeung C. A., Tsang T. Y., Yau E., Luo K., Wu P., et al. (2012). Clitocine reversal of P-glycoprotein associated multi-drug resistance through down-regulation of transcription factor NF-κB in R-HepG2 cell line. PloS One 7, e40720. 10.1371/journal.pone.0040720 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suzuki T., Fukazawa N., San-nohe K., Sato W., Yano O., Tsuruo T. (1997). Structure– activity relationship of newly synthesized quinoline derivatives for reversal of multidrug resistance in cancer. J. Med. Chem. 40, 2047–2052. 10.1021/jm960869l [DOI] [PubMed] [Google Scholar]
- Suzuki H., Tanabe H., Mizukami H., Inoue M. (2010). Selective regulation of multidrug resistance protein in vascular smooth muscle cells by the isoquinoline alkaloid coptisine. Biol. Pharm. Bull. 33, 677–682. 10.1248/bpb.33.677 [DOI] [PubMed] [Google Scholar]
- Syed S. B., Coumar M. S. (2016). P-Glycoprotein mediated multidrug resistance reversal by phytochemicals: a review of SAR & future perspective for drug design. Curr. Top. Med. Chem. 16, 2484–2508. 10.2174/1568026616666160212123814 [DOI] [PubMed] [Google Scholar]
- Syed S. B., Arya H., Fu I. H., Yeh T. K., Periyasamy L., Hsieh H. P., et al. (2017). Targeting P-glycoprotein: Investigation of piperine analogs for overcoming drug resistance in cancer. Sci. Rep. 7, 1–18. 10.1038/s41598-017-08062-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tanaka K., Iwamoto S., Gon G., Nohara T., Iwamoto M., Tanigawa N. (2000). Expression of survivin and its relationship to loss of apoptosis in breast carcinomas. Clin. Canc. Res. 6, 127–134. [PubMed] [Google Scholar]
- Tang X., Bi H., Feng J., Cao J. (2005). Effect of curcumin on multidrug resistance in resistant human gastric carcinoma cell line SGC7901/VCR. Acta Pharmacol. Sin. 26, 1009–1016. 10.1111/j.1745-7254.2005.00149.x [DOI] [PubMed] [Google Scholar]
- Teng Y. N., Sheu M. J., Hsieh Y. W., Wang R. Y., Chiang Y. C., Hung C. C. (2016). β-carotene reverses multidrug resistant cancer cells by selectively modulating human P-glycoprotein function. Phytomedicine 23, 316–323. 10.1016/j.phymed.2016.01.008 [DOI] [PubMed] [Google Scholar]
- Tiwary B. K., Pradhan K., Nanda A. K., Chakraborty R. (2015). Implication of quinazoline-4(3H)-ones in medicinal chemistry: a brief review. J. Chem. Biol. Ther. 1, 104. 10.4172/2572-0406.1000104 [DOI] [Google Scholar]
- To K. K., Wu X., Yin C., Chai S., Yao S., Kadioglu O. (2017). Reversal of multidrug resistance by Marsdenia tenacissima and its main active ingredients polyoxypregnanes. J. Ethnopharmacol. 203, 110–119. 10.1016/j.jep.2017.03.051 [DOI] [PubMed] [Google Scholar]
- Trock B. J., Leonessa F., Clarke R. (1997). Multidrug resistance in breast cancer: A Meta-analysis of MDR1/gp170 expression and its possible functional significance. J. Natl. Canc. Inst. 89, 917–931. 10.1093/jnci/89.13.917 [DOI] [PubMed] [Google Scholar]
- Um Y., Cho S., Woo H. B., Kim Y. K., Kim H., Ham J., et al. (2008). Synthesis of curcumin mimics with multidrug resistance reversal activities. Bioorg. Med. Chem. 16, 3608–3615. 10.1016/j.bmc.2008.02.012 [DOI] [PubMed] [Google Scholar]
- Umsumarng S., Pitchakarn P., Yodkeeree S., Punfa W., Mapoung S., Ramli R. A. (2017). Modulation of P-glycoprotein by Stemona alkaloids in human multidrug resistance leukemic cells and structural relationships. Phytomedicine 34, 182–190. 10.1016/j.phymed.2017.08.004 [DOI] [PubMed] [Google Scholar]
- van Wyk B. E., Wink M. (2017). Medicinal Plants of the World (Wallingford, UK: CABI; ). [Google Scholar]
- van Zanden J. J., Wortelboer H. M., Bijlsma S., Punt A., Usta M., Bladeren v., et al. (2005). Quantitative structure activity relationship studies on the flavonoid mediated inhibition of multidrug resistance proteins 1 and 2. Biochem. Pharmacol. 69, 699–708. 10.1016/j.bcp.2004.11.002 [DOI] [PubMed] [Google Scholar]
- Vasas A., Sulyok E., Rédei D., Forgo P., Szabó P., Zupkó I., et al. (2011). Jatrophane diterpenes from Euphorbia esula as antiproliferative agents and potent chemosensitizers to overcome multidrug resistance. J. Nat. Prod. 74, 1453–1461. 10.1021/np200202h [DOI] [PubMed] [Google Scholar]
- Wan C. K., Zhu G. Y., Shen X. L., Chattopadhyay A., Dey S., Fong W. F. (2006). Gomisin A alters substrate interaction and reverses P-glycoprotein-mediated multidrug resistance in HepG2-DR cells. Biochem. Pharmacol. 72, 824–837. 10.1016/j.bcp.2006.06.036 [DOI] [PubMed] [Google Scholar]
- Wandel C., Kim R. B., Kajiji S., Guengerich F. P., Wilkinson G. R., Wood A. J. (1999). P-glycoprotein and cytochrome P-450 3A inhibition: dissociation of inhibitory potencies. Canc. Res. 59, 3944–3948. [PubMed] [Google Scholar]
- Wang Y., Cabral F. (2005). Paclitaxel resistance in cells with reduced β-tubulin. Biochim. Biophys. Acta Mol. Cell Res. 1744, 245–255. 10.1016/j.bbamcr.2004.12.003 [DOI] [PubMed] [Google Scholar]
- Wang E. J., Barecki-Roach M., Johnson W. W. (2002). Elevation of P-glycoprotein function by a catechin in green tea. Biochem. Biophys. Res. Commun. 297, 412–418. 10.1016/S0006-291X(02)02219-2 [DOI] [PubMed] [Google Scholar]
- Wang C., Zhang J. X., Shen X. L., Wan C. K., Tse A. K. W., Fong W. F. (2004). Reversal of P-glycoprotein-mediated multidrug resistance by Alisol B 23-acetate. Biochem. Pharmacol. 68, 843–855. 10.1016/j.bcp.2004.05.021 [DOI] [PubMed] [Google Scholar]
- Wang X. B., Wang S. S., Zhang Q. F., Liu M., Li H. L., Liu Y., et al. (2010). Inhibition of tetramethylpyrazine on P-gp, MRP2, MRP3 and MRP5 in multidrug resistant human hepatocellular carcinoma cells. Oncol. Rep. 23, 211–215. 10.3892/or_00000625 [DOI] [PubMed] [Google Scholar]
- Wang S., Lei T., Zhang M. (2016). The reversal effect and its mechanisms of tetramethylpyrazine on multidrug resistance in human bladder cancer. PloS One 11, e0157759. 10.1371/journal.pone.0157759 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang X., Zhang H., Chen X. (2019). Drug resistance and combating drug resistance in cancer. Canc. Drug Resist. 2, 141–160. 10.20517/cdr.2019.10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wesołowska O., Hendrich A. B., Łania-Pietrzak B., Wiśniewski J., Molnár J., Ocsovszki I., et al. (2009). Perturbation of the lipid phase of a membrane is not involved in the modulation of MRP1 transport activity by flavonoids. Cell. Mol. Biol. Lett. 14, 199. 10.2478/s11658-008-0044-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wink M., van Wyk B. E. (2008). Mind-Altering and Poisonous Plants of the World (Portland, USA: Timber Press; ). [Google Scholar]
- Wink M., Ashour M. L., El-Readi M. Z. (2012). Secondary metabolites inhibiting ABC transporters and reversing resistance of cancer cells and fungi to cytotoxic and antimicrobial agents. Front. Microbiol. 3, 130. 10.3389/fmicb.2012.00130 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wink M. (2000). “Interference of alkaloids with neuroreceptors and ion channels,” in Studies in Natural Products Chemistry. Ed. Ata-ur- R. (Amsterdam, Netherlands: Elsevier; ), 3–122. [Google Scholar]
- Wink M. (2007). “Molecular modes of action of cytotoxic alkaloids: from DNA intercalation, spindle poisoning, topoisomerase inhibition to apoptosis and multiple drug resistance,” in The Alkaloids: Chemistry and Biology, vol. 64 (San Diego, USA: Academic Press; ), 1–47. [DOI] [PubMed] [Google Scholar]
- Wink M. (2015). Modes of action of herbal medicines and plant secondary metabolites. Medicines 2, 251–286. 10.3390/medicines2030251 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wink M. (2020). Evolution of the angiosperms and co-evolution of secondary metabolites, especially of Alkaloids. Reference Series in Phytochemistry. Co-Evolution of Secondary Metabolites (Cham, Switzerland: Springer Nature; ). [Google Scholar]
- Wong I. L., Wang B. C., Yuan J., Duan L. X., Liu Z., Liu T., et al. (2015). Potent and nontoxic chemosensitizer of P-glycoprotein-mediated multidrug resistance in cancer: synthesis and evaluation of methylated epigallocatechin, gallocatechin, and dihydromyricetin derivatives. J. Med. Chem. 58, 4529–4549. 10.1021/acs.jmedchem.5b00085 [DOI] [PubMed] [Google Scholar]
- Wongrattanakamon P., Lee V. S., Nimmanpipug P., Sirithunyalug B., Chansakaow S., Jiranusornkul S. (2017). Insight into the molecular mechanism of p-glycoprotein mediated drug toxicity induced by bioflavonoids: An integrated computational approach. Toxicol. Mech. Meth. 27, 253–271. 10.1080/15376516.2016.1273428 [DOI] [PubMed] [Google Scholar]
- Wu J. Y. C., Fong W. F., Zhang J. X., Leung C. H., Kwong H. L., Yang M. S., et al. (2003). Reversal of multidrug resistance in cancer cells by pyranocoumarins isolated from Radix peucedani. Eur. J. Pharmacol. 473, 9–17. 10.1016/S0014-2999(03)01946-0 [DOI] [PubMed] [Google Scholar]
- Xu D., Lu Q., Hu X. (2006). Down-regulation of P-glycoprotein expression in MDR breast cancer cell MCF-7/ADR by honokiol. Canc. Lett. 243, 274–280. 10.1016/j.canlet.2005.11.031 [DOI] [PubMed] [Google Scholar]
- Ye J., Zheng Y., Liu D. (2009). Reversal effect and its mechanism of ampelopsin on multidrug resistance in K562/ADR cells. China J. Chin. Materia Med. 34, 761–765. [PubMed] [Google Scholar]
- Yoo H. H., Lee M., Chung H. J., Lee S. K., Kim D. H. (2007. a). Effects of diosmin, a flavonoid glycoside in citrus fruits, on P-glycoprotein-mediated drug efflux in human intestinal Caco-2 cells. J. Agr. Food Chem. 55, 7620–7625. 10.1021/jf070893f [DOI] [PubMed] [Google Scholar]
- Yoo H. H., Lee M., Lee M. W., Lim S. Y., Shin J., Kim D. H. (2007. b). Effects of Schisandra lignans on P-glycoprotein-mediated drug efflux in human intestinal Caco-2 cells. Planta Med. 53, 444–450. 10.1055/s-2007-967178 [DOI] [PubMed] [Google Scholar]
- Yoshida N., Takagi A., Kitazawa H., Kawakami J., Adachi I. (2005). Inhibition of P-glycoprotein-mediated transport by extracts of and monoterpenoids contained in Zanthoxyli fructus. Toxicol. Appl. Pharmacol. 209, 167–173. 10.1016/j.taap.2005.04.001 [DOI] [PubMed] [Google Scholar]
- You M., Ma X., Mukherjee R., Farnsworth N. R., Cordell G. A., Kinghorn A. D., et al. (1994). Indole alkaloids from Peschiera laeta that enhance vinblastine-mediated cytotoxicity with multidrug-resistant cells. J. Nat. Prod. 57, 1517–1522. 10.1021/np50113a007 [DOI] [PubMed] [Google Scholar]
- You M., Wickramaratne D. M., Silva G. L., Chai H., Chagwedera T. E., Farnsworth N. R., et al. (1995). (-)-Roemerine, an aporphine alkaloid from Annona senegalensis that reverses the multidrug-resistance phenotype with cultured cells. J. Nat. Prod. 58, 598–604. 10.1021/np50118a021 [DOI] [PubMed] [Google Scholar]
- Zhang S., Morris M. E. (2003). Effects of the flavonoids biochanin A, morin, phloretin, and silymarin on P-glycoprotein-mediated transport. J. Pharmacol. Exp. Therapeut. 304, 1258–1267. 10.1124/jpet.102.044412 [DOI] [PubMed] [Google Scholar]
- Zhang C. C., Yang J. M., White E., Murphy M., Levine A., Hait W. N. (1998). The role of MAP4 expression in the sensitivity to paclitaxel and resistance to vinca alkaloids in p53 mutant cells. Oncogene 16, 1617–1624. 10.1038/sj.onc.1201658 [DOI] [PubMed] [Google Scholar]
- Zhang S., Yang X., Morris M. E. (2004). Combined effects of multiple flavonoids on breast cancer resistance protein (ABCG2)-mediated transport. Pharm. Res. 21, 1263–1273. 10.1023/B:PHAM.0000033015.84146.4c [DOI] [PubMed] [Google Scholar]
- Zhang S., Wang X., Sagawa K., Morris M. E. (2005). Flavonoids chrysin and benzoflavone, potent breast cancer resistance protein inhibitors, have no significant effect on topotecan pharmacokinetics in rats or mdr1a/1b (–/–) mice. Drug Metabol. Dispos. 33, 341–348. 10.1124/dmd.104.002501 [DOI] [PubMed] [Google Scholar]
- Zhang Y., Liu X., Zuo T., Liu Y., Zhang J. H. (2012). Tetramethylpyrazine reverses multidrug resistance in breast cancer cells through regulating the expression and function of P-glycoprotein. Med. Oncol. 29, 534–538. 10.1007/s12032-011-9950-8 [DOI] [PubMed] [Google Scholar]
- Zhou N., Li J., Li T., Chen G., Zhang Z., Si Z. (2017). Matrine-induced apoptosis in Hep3B cells via the inhibition of MDM2. Mol. Med. Rep. 15, 442–450. 10.3892/mmr.2016.5999 [DOI] [PubMed] [Google Scholar]
- Zhu X., Zhang X., Ma G., Yan J., Wang H., Yang Q. (2011). Transport characteristics of tryptanthrin and its inhibitory effect on P-gp and MRP2 in Caco-2 cells. J. Pharm. Pharmaceut. Sci. 14, 325–335. 10.18433/J3501W [DOI] [PubMed] [Google Scholar]
- Zhu J., Wang R., Lou L., Li W., Tang G., Bu X., et al. (2016). Jatrophane diterpenoids as modulators of P-glycoprotein-dependent multidrug resistance (MDR): advances of structure–activity relationships and discovery of promising MDR reversal agents. J. Med. Chem. 59, 6353–6369. 10.1021/acs.jmedchem.6b00605 [DOI] [PubMed] [Google Scholar]