Lindsay K.
Caesar
and
Nadja B.
Cech
*
Department of Chemistry & Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina, USA. E-mail: nadja_cech@uncg.edu
First published on 12th June 2019
Covering: 2000 to 2019
According to a 2012 survey from the Centers for Disease Control and Prevention, approximately 18% of the U.S. population uses natural products (including plant-based or botanical preparations) for treatment or prevention of disease. The use of plant-based medicines is even more prevalent in developing countries, where for many they constitute the primary health care modality. Proponents of the medicinal use of natural product mixtures often claim that they are more effective than purified compounds due to beneficial “synergistic” interactions. A less-discussed phenomenon, antagonism, in which effects of active constituents are masked by other compounds in a complex mixture, also occurs in natural product mixtures. Synergy and antagonism are notoriously difficult to study in a rigorous fashion, particularly given that natural products chemistry research methodology is typically devoted to reducing complexity and identifying single active constituents for drug development. This report represents a critical review with commentary about the current state of the scientific literature as it relates to studying combination effects (including both synergy and antagonism) in natural product extracts. We provide particular emphasis on analytical and Big Data approaches for identifying synergistic or antagonistic combinations and elucidating the mechanisms that underlie their interactions. Specific case studies of botanicals in which synergistic interactions have been documented are also discussed. The topic of synergy is important given that consumer use of botanical natural products and associated safety concerns continue to garner attention by the public and the media. Guidance by the natural products community is needed to provide strategies for effective evaluation of safety and toxicity of botanical mixtures and to drive discovery in botanical natural product research.
Despite centuries of use, the activity of botanical medicines is only partially understood, and for most natural products on the market, there is a lack of knowledge as to which constituents are responsible for the purported biological activity. Scientific investigation of botanical natural products is challenging because of their immense complexity and variability.7–9 Natural products chemistry efforts are typically devoted to reducing complexity and identifying single “active” constituents for drug development. However, given that complex plant extracts, and not single molecules, are often administered for medicinal purposes, interactions between constituents could be of great importance.
Understanding how mixtures work in concert to achieve a given biological effect may address the ever-increasing threat of disease resistance. Indeed, many diseases are not regulated by a single molecular target, but often have a multi-factorial causality.7,9 It has been shown in numerous studies that disease resistance is less likely to occur against a combination of compounds than to single active constituents.8,10 Plants have evolved over millennia to address the multifactorial nature of disease pathogenesis by targeting pathogens through the combined action of structurally and functionally diverse constituents.7,11 As such, complex natural product mixtures offer an important resource for drug development, and to ensure future success in natural products research, understanding interactions within and between the constituents of natural product mixtures is paramount.
Pharmacological investigations into combination effects can be examined at the level of the molecular targets, disease pathways, cellular processes, and patient responses.12 As such, in vitro, in vivo, pre-clinical, and clinical research can all provide valuable insight into combination effects. Considerable progress has been made in the clinical arena in terms of investigating drug synergy, reviewed extensively in several publications.12–15 While much research is being conducted in this realm, this review will focus primarily on methodology to interpret combination effects using molecular and cellular methods.
Botanical extracts may contain hundreds or even thousands of individual constituents at varying abundance16 (Fig. 1) and identifying the compounds responsible for a given biological effect represents a significant challenge. Too often, it is assumed that the behaviour of a mixture can be described by the presence of just a few known constituents. However, a number of studies have shown that the overall activity of botanical extracts can result from mixtures of compounds with synergistic, additive, or antagonistic activity,9,17–20 and those who work in the field of botanical natural products research will be quick to admit that it is very often the case that isolation efforts on a botanical extract fail because activity is lost upon fractionation.9,17,20 While there are multiple possible explanations for this failure (including irreversible adsorption of compounds to the column packing),21 it is certainly true that in some cases loss of activity occurs because multiple constituents are required to observe the biological effect. Many investigators recognize the multi-factorial nature of botanical medicines. However, research methodology as applied to botanical mixtures still tends, in most cases, either to take a reductionist approach (focusing on just one or two “marker compounds”) or to ignore the question of chemical composition altogether, testing the biological effects of complex mixtures for which active constituents are unknown. The problem in the latter case is that results tend to be difficult both to interpret and to reproduce. Herein, we seek to provide an overview of the methodology that currently exists to understand combination effects within complex mixtures. We will highlight existing technologies for studying combination effects, placing particular emphasis on – Omics technologies and other Big Data approaches that have developed significantly in the last several years. We aim to provide practical advice to investigators seeking to comprehensively evaluate the constituents and mechanisms responsible for the biological activity of botanical mixtures.
To overcome some of these challenges, many researchers work with in vitro systems instead. However, many cell-free, high-throughput assays that search for molecular targets do not accurately model the biology of an intact cell, making the discovery of relevant combination effects unfeasible.32 As such, cell-based assays can be employed that strike a balance between efficiency and preservation of molecular pathway interactions.33 Many useful cellular systems for identifying combination effects in vitro have been discussed in a recent publication by Pemovska et al.12 In addition to choosing relevant cellular systems for conducting biological testing, it is important to mimic physiological conditions in the assay itself. Indeed, most media used to grow cells for biological testing do not mimic physiological conditions, influencing the metabolism and phenotypic response of the cells under study.34 Similarly, biological assay conditions can result in “dynamic residual complexity,” in which the sample is subjected to an environmentally-induced chemical change, making interpretation of results challenging.35 In their recent publication, Vande Voorde et al.34 illustrated that the utilization of a complex culture medium designed to mimic the physiological environment of cancer cells prevented the formation of unwanted phenotypic artifacts and improved the translatability between in vitro assay results and in vivo tumor models. The utilization of physiologically relevant media also improves the likelihood that components that elicit a biological response during biological testing will be soluble and stable in a biological system, facilitating identification of active constituents. Primary tissue assays comprised of multiple cell types, such as those used to screen drug combinations for anti-inflammatory activity in mixed cultures of lymphocytes, can also be used to reveal combination effects that work through multi-target mechanisms.32
When screening for biological activity in vitro, however, researchers should be aware of potential false-positive results originating from pan-assay interference compounds, commonly referred to as PAINS, which are often identified as hits in biological screens.35 These false positive results may occur through a variety of mechanisms, including fluorescence quenching, aggregation effects, chemical reactivity, oxidation/reduction, membrane disruption, and residual complexity.35 Synergy results are often identified in aqueous media due to aggregation effects, which can be minimized by the addition of detergent to the media.36 While the promiscuous nature of PAINS compounds may in some cases be cause for concern, numerous examples of clinical drugs contain substructures that fall into this category.37 For example, many quinone-based drugs have been approved by the FDA for their antineoplastic, immunosuppressant, and antiprotozoal activities.37 As such, biological assay results should be seen as hypothesis-generating tools, and further verification is required to identify true leads (or to eliminate “PAINS” compounds from consideration).
In addition to carefully choosing the biological system to study for combination effects, data enabling the efficient comparison of a drug combination to agents in isolation must be gathered.8,32 Combination effects including synergy and antagonism can occur over a broad range of concentrations, so various ratios of the samples under study must be tested.8,32,33,38–41 Numerous methodologies have been developed to acquire data to discover combination effects in vitro, including checkerboard assays and time-kill methods, many of which are quite labor- and material-intensive.8,32,42 One of the simplest methods for identifying potential combination effects is through testing samples alone and in combination, and determining if the combined effect of the samples is greater, equal, or less than the expected sum of the two samples in isolation. Although simple, assays employing this approach cannot claim synergy without further study because they lack the range of concentration combinations required to fully assess combination effects, and should be used only to prioritize samples for more in-depth studies.43 These in-depth studies can be achieved using a dose–response matrix design,33 also known as a checkerboard assay, in which a series of dose–response curves using different dose combinations of the agents under study are acquired and compared.8,32
In addition to concentration-based approaches to evaluate combination effects, time-based approaches have also been developed and applied to identify antimicrobial synergy and to describe the relationship between bactericidal activity and sample concentration.44 This method involves exposing a selected pathogen to an inhibitor (or combination of inhibitors), sampling cultures at regular time intervals, serially diluting and incubating aliquots, and comparing the colony forming units produced. The resulting dose–response curve can be used to define additive, synergistic, and antagonistic effects.44 Importantly, several of these methods have been compared using the same datasets,38–41 revealing a lack of consistency between conclusions met using these approaches.38–40,42,45 Not only do in vitro tests often result in conflicting results, but it is very often the case that reproducible hits in vitro lack efficacy in vivo.30 Because of this inconsistency, preliminary screening efforts should be used to prioritize candidates with potential synergy but should not be used to unequivocally define combination effects.
Despite the existence of numerous reference models, the general isobole equation, based on the assumptions of the Loewe additivity principle, remains the most popular for studying combination effects.8,9,27–29,55 As described elsewhere, an isobole, or an “isobologram,” is a graphical representation of the combination effects between two samples.8,9,27–29,55 The axes of the plot represent the doses of individual agents, and the points plotted indicate the combination of concentrations of the two treatments required to reach a particular fixed effect (i.e. 50% inhibition of cell growth).8 If the two samples have no interaction, the line joining the axes will be a straight line. Synergy will result in a concave curve, and antagonism will result in a convex curve (Fig. 2).8,9,27–29 In a recent publication, Lederer et al.55 scrutinized the implicit assumptions of the Loewe additivity model (and with it the general isobole equation), and found that the consistency of the model only holds if the two samples under study do not differ in the slopes nor the maximal effects of their dose–response curves.55 In cases where one sample reaches an effect that cannot be reached by the other sample, the Loewe additivity consistency condition is violated.54,56 To overcome this limitation with the Loewe additivity consistency condition, Lederer et al.55 developed an adaptation of the general isobole equation termed the explicit mean equation. The explicit mean equation is equivalent to the isobole equation in cases where the two samples meet the Loewe additivity consistency condition and is capable of identifying combination effects even if this condition is violated. In a follow up study, Lederer et al.46 compared six models built on either Loewe additivity or Bliss independence principles using existing, high-throughput datasets57,58 and found that Loewe additivity models performed better than Bliss independence at separating synergy relationships from other combination effects and that the explicit mean equation was the overall best performing model.46
In recent years, variants of the Loewe additivity model and the Bliss independence model have been developed.56,59–63 However, because the expected responses from these different models are often disparate,27,46 it is challenging to draw biological conclusions from the resulting data. In some instances, combination effects have been identified as synergistic by one model but antagonistic by another.57 As such, researchers should be clear about which model they have chosen to adopt, as stated in the Saariselkä agreement.64 Tang et al.26 have expanded upon this suggestion and have proposed the use of terminology that incorporates results from both Bliss independence and Loewe additivity models. The authors argue that the level of consistency between models should be used to designate the degree of synergy or antagonism. For example, if both models identify a given interaction as synergistic, that interaction should be considered “strong synergy,” and if the combination is identified as synergistic by one model only, it should be considered “weak synergy”.26 By utilizing both models, this proposal minimizes the incorporation of bias into predictions and provides more informative definitions for the combination effects described. While in principle this proposal makes sense, it also relies on the assumption that the models are equally valid. While Loewe additivity models have been shown to perform better than Bliss independence models on numerous occasions,46 Russ and Kishony65 found that the Bliss independence models are more consistent when interactions between more than two samples are evaluated. As such, the use of any synergy model should be seen only as a hypothesis-generating tool to prioritize potential interaction effects for further study. Indisputable definitions of synergy and antagonism remain elusive, and a wider agreement on the terminology used for interaction assessment is still required to standardize future research initiatives.
(1) |
In this equation, A and B represent the samples under study, IC50A and IC50B represent the concentrations of A or B in isolation to reach 50% inhibition, [A] is the IC50 of A in the presence of B, and [B] is the IC50 of B in the presence of A. Notably, any fixed effect can be used to calculate ∑FIC indices, but IC50 values are perhaps the most common metric.
Despite the popularity of this method, the interpretation of ∑FIC scores for defining combination effects varies considerably from author to author. In their recent publication, van Vuuren and Viljoen8 provide an excellent commentary on ∑FIC score interpretation, improving upon the earliest interpretations proposed by Berenbaum in which synergistic interactions were considered to be any value below one, additive/indifferent interactions focused on one, and antagonistic interactions above one.27 However, because of the inconsistency across null reference models, and because fixed effects can often be placed within a three-dilution range using in vitro assays,68 a more conservative approach is warranted. Taking this into consideration, van Vuuren and Viljoen8 and the authors of this review suggest that synergistic interactions be defined as interactions having ∑FIC ≤ 0.5, additive interactions range from 0.5 to 1.0, non-interactive effects range from 1.0 to 4.0, and antagonistic effects fall above 4.0 (Table 1).
Combination effect | ∑FIC range |
---|---|
Synergy | ∑FIC ≤ 0.5 |
Additivity | 0.5 < ∑FIC ≤ 1.0 |
Indifference | 1.0 < ∑FIC ≤ 4.0 |
Antagonism | 4.0 < ∑FIC |
Despite its popularity, the ∑FIC index, like the isobologram upon which it is based, is insufficient to effectively capture the combination effects that may occur across multiple dose regions.46,47 An inherent limitation of the ∑FIC index is the focus on a single interaction parameter. In a recent publication, Lederer et al.46 compared multiple synergy measurements and found that the “lack of fit” model,69 where synergy scores are defined by the volume spanned between the null reference model and the measured response, performed better than parametric models in its ability to identify synergistic effects.46
Similarly, Yadav et al. developed a score that enables the use of an interaction landscape over the full dose–response matrix to identify combination effects across multiple dosages and response levels.47 Rather than relying on a single parameter such as the IC50 measurement, the delta-score utilized in this study was calculated by assessing changes in both the shape parameter and the midpoint of each dose–response curve for individual samples and combinations thereof. The delta scores were visualized using a response surface plot to visualize the combination effect landscape over all tested dosage combinations, enabling identification of potency changes and differences in combination effects even within the same sample pair (Fig. 3). There appears to be value in using these different methods to explore synergy; however, these approaches have not yet been applied to understand synergy in complex natural products and discussion of their merit for this purpose remains hypothetical. Despite the aforementioned limitations, isobole analysis and the ∑FIC index have found the widest utility in natural products research.8
Fig. 3 Example of synergistic (top) and antagonistic (bottom) interaction landscapes using delta scores (δ) calculated with the zero interaction potency model of compounds in combination with ibrutinib, an approved anti-cancer drug targeting Bruton's tyrosine kinase. (A) Interaction map between anti-cancer activity of ispinesib (a selective kinesin spindle protein inhibitor) and ibrutinib. Average delta across the dose–response matrix (Δ) is 17.596, indicative of overall synergy. (B) Interaction map between canertinib (an epidermal growth factor receptor family inhibitor) and ibrutinib. The Δ value is −14.038, indicative of overall antagonism. Figure is reprinted with permission from Yadav et al. 2015.47 |
In addition to artemisinin, there are approximately 30 other flavonoids and sesquiterpenes within A. annua, some of which have minor anti-plasmodial activities.86 As might be expected, since botanical preparations are multi-factorial rather than monospecific in nature, both in vitro and in vivo studies evaluating the activity of A. annua extracts have found that the amount of artemisinin in the extracts does not fully explain the extract's efficacy against P. falciparum parasites.87,88 Indeed, various combination therapies including artemisinin and its derivatives are utilized as antimalarial treatments.89,90 In a recent study, Suberu et al.91 aimed to identify the compounds within A. annua tea extract contributing to its anti-plasmodial efficacy. Building upon the work of previous studies which found that several flavonoids potentiated the activity of artemisinin against P. falciparum,92,93 Suberu et al.91 tested the tea extract, purified standards from the extract, and various combinations of artemisinin with purified compounds against both chloroquine-sensitive and chloroquine-resistant strains of P. falciparum. Interestingly, the type of combination effect observed, whether it be synergistic, additive, or antagonistic, often differed depending on the dosage of the combined constituents and/or the resistance profile of the parasite under analysis.91
Using isobologram analysis and calculating ∑FIC indices, Suberu et al.91 found several compounds that enhanced or antagonized the activity of artemisinin against P. falciparum. Two compounds that contained anti-plasmodial activity, 9-epi-artemisinin and artemisitene, were found to antagonize the efficacy of artemisinin against both chloroquine-sensitive and chloroquine-resistant strains. Although the mechanism by which these compounds antagonize artemisinin's activity is unknown, it is reasonable to assume these compounds, which have only minor structural differences, compete for the same molecular target, reducing the overall efficacy of the compounds in combination.91 Several additional compounds contained within the extract, however, did not demonstrate the same combination effect at all concentrations tested. For example, 3-caffeoylquinic acid showed a summation effect in combination with artemisinin at a ratio of 1:3 (artemisinin to 3-caffeoylquinic acid) when tested against the chloroquine-sensitive strain, but at higher combination ratios (1:10–100), synergistic interactions were observed. Similarly, casticin, which possessed antagonistic activity at the 1:3 ratio, has been reported to be synergistic in other studies using higher combination ratios (1:10–1000).92,93 The reason for this discrepancy is unknown, but it is possible that these compounds act as either anti-oxidant or pro-oxidant species depending on the dosage level.94,95 When combined at a low concentration with artemisinin, they may have counteracted artemisinin activity through anti-oxidative interaction, minimizing the oxidative stress resulting from the reactive oxygen species formed through artemisinin's activity, while at higher concentrations they were pro-oxidative, increasing the oxidative stress and leading to increased efficacy of artemisinin.91
Other compounds, including rosmarinic acid and arteannuin B, showed differential combination effects when tested against sensitive and resistant strains of P. falciparum. Rosmarinic acid was synergistic against the sensitive strain, but showed antagonistic activity in the resistant strain.91 Similarly, arteannuin B had an additive/indifferent interaction in the chloroquine sensitive strain, but a synergistic interaction with the resistant strain, leading to a three-fold improvement in artemisinin's activity.91 Because arteannuin B selectively potentiates the activity of artemisinin in the chloroquine-resistant strain, it likely targets the parasite's chloroquine resistance mechanism, illustrating the promise of combination treatments not only for developing therapeutics against drug-resistant pathogens, but also for providing insight into the mechanisms by which parasites gain resistance as a whole.
It is important to note that Suberu et al. chose somewhat liberal ranges for the ∑FIC indices used to define their combination effects,91 and other researchers, depending on the models chosen, may have categorized some of the synergistic and antagonistic interactions as “additive” or “indifferent”.8 Even if one were to re-categorize interactions based on conservative estimates, however, all three types of combination effects (synergy, additivity, and antagonism) were witnessed during the course of this study. While the specific categorizations of synergy, additivity, and antagonism chosen by Suberu et al. may be disputed, it is clear that the nature of combination effects did often change depending on both the dosage and the parasite strain under study.91
The constituents responsible for the activity of Echinacea purpurea (L.) Moench (Asteraceae) extracts and the mechanisms by which these constituents exert their purported beneficial effects have been studied extensively. Early research on Echinacea attributed its purported health benefits to its ability to “activate” or “stimulate” immune cells. These findings were based upon early work by Wagner and co-workers, in which isolated Echinacea polysaccharides were observed to stimulate phagocytosis and induce TNF-alpha secretion by macrophages.100 Later research demonstrated that much of the immunostimulatory activity originally attributed to Echinacea polysaccharides could instead be linked to the lipopolysaccharides and lipoproteins. These lipoproteins and lipopolysaccharides are components of bacterial cell walls, and can be attributed to the presence of bacterial endophytes, bacteria living asymptomatically within the Echinacea plant tissues.101–104 Even very minute quantities of certain lipoproteins and lipopolysaccharides induce pronounced immunostimulatory effects in macrophages, so the presence of these compounds as contaminants can confound in vitro assay data.
An alternative narrative about the immunomodulatory activity of Echinacea preparations focused on alkylamide constituents. Contrary to the research on polysaccharides, lipoproteins, and lipopolysaccharides, these alkylamides were observed to suppress the production of pro-inflammatory cytokines by macrophages.105–108 Such activity could translate to a beneficial anti-inflammatory effect in vivo. The apparently contradictory activity of various classes of compounds, both isolated from Echinacea, suggested the possibility that the activity of some constituents might be masked by others in the context of complex Echinacea extracts. This was shown in a study by Todd et al.,104 in which complex E. purpurea extracts possessed little to no cytokine-suppressive activity, but could be separated to produce sub-fractions with opposing effects. Some fractions, those containing alkylamides, suppressed cytokine and chemokine production by macrophages, while others, those containing lipopolysaccharides, induced cytokine production. Thus, it was demonstrated that lipopolysaccharides (and likely other compounds of bacterial origin) masked the anti-inflammatory effect of complex Echinacea preparations, effectively acting as antagonists. It was not until these fractions were separated that the individual activities of the various constituents could be observed.
Salvia miltiorrhiza Bunge (Lamiaceae) and Pueraria lobata (Willd.) Ohwi (Fabaceae), known as Danshen and Gegen, respectively, are often combined to treat coronary heart disease.110 In a recent study, Danshen and Gegen were tested both alone and in combination in order to confirm the presence of anti-atherogenic effects and to identify potential combination effects between herbal constituents. Biological effects of the three extracts (Danshen and Gegen alone and in combination) were tested for their anti-inflammatory, anti-foam cell formation, and anti-vascular smooth muscle cell (vSMC) proliferation effects.110 The biological assay results were evaluated using both fixed-ratio experimental design and fractional inhibitory concentration indices (alternatively named “combination indices” in this publication). The Danshen–Gegen combination was characterized as synergistic in the anti-inflammation assay, additive in the foam cell formation assay, and antagonistic in the vSMC proliferation assay, with ∑FIC indices of 0.75, 1.03, and 2.02, respectively.110 When categorizing these combination effects, however, the authors of this study chose quite lenient values. Using the recommended values in this review and others,8 the combination effects witnessed in these three assays would be re-categorized as additive (for the anti-inflammation assay) and indifferent (for the foam-cell formation and vSMC proliferation assays). Nonetheless, this study highlights the applicability of multi-herb formulas to treat disease, and provides rationale for the combination of Danshen and Gegen for the treatment of atherogenesis.110
Oftentimes, disease targets are able to counteract the therapeutic effect of an active metabolite, resulting in its reduced efficacy.76 One type of pharmacodynamic synergism involves “anti-counteractive action” in which a synergistic compound binds to an anti-target, effectively inhibiting the disease target from counteracting the therapeutic effect of the active constituent.76 Pharmacodynamic synergy may also occur through complementary actions, in which synergists in a mixture interact with multiple points of a given pathway, resulting in positive regulation of a process affecting the drug target or in the negative regulation of competing mechanisms. Through the selective variation of target activity and expression through complementary actions, pharmacodynamic synergists can both augment beneficial effects of treatments and reduce adverse effects of the disease.76 For example, Ginkgo biloba L. (Ginkgoaceae) has been shown in numerous studies to have synergistic neuroprotective effects both in vivo and in vitro by inhibiting the formation of free radicals, scavenging reactive oxygen species, regulating gene expression of mitochondrial targets, and reducing excessive stimulation of nerve cells by neurotransmitters.66,120
Absorption of active constituents can be improved through a variety of mechanisms, including the inhibition of drug exporters such as P-glycoproteins.111,124,125 Additionally, transport barriers may be disrupted or their recovery delayed, improving permeability of active constituents into target cells.76 For example, the absorption of baicalin, a constituent of the plant Scutellaria baicalensis Georgi (Lamiaceae), is synergistically enhanced by the addition of both coumarins and volatile oils from the botanical Angelica dahurica Bentham et Hooker (Apiaceae), likely by affecting transport systems independent of P-glycoproteins.126 Pharmacokinetic synergy also results from constituents that inhibit enzymes that convert drugs into excretable or inactive forms, or that activate enzymes that convert pro-drugs into active forms.7,76
Fig. 4 Bacterial resistance mechanisms that could be targeted with combination therapy enabling re-sensitization of resistant organisms to existing antibiotics. |
Bacterial resistance to beta-lactam antibiotics is achieved by the development of beta-lactamase enzymes that cleave the antibiotics into inactive forms.137 This resistance mechanism can be overcome by combining beta-lactam antibiotics with beta-lactamase inhibitors. In a recent study, Catteau et al.138 found that a dichloromethane extract of shea butter tree leaves (Vitellaria paradoxa C.F. Gaertn. (Saptoaceae)) synergized the activity of ampicillin, oxacillin, and nafcillin against methicillin-resistant Staphylococcus aureus by targeting PBP2a +/− beta-lactamase enzymes. V. paradoxa constituents ursolic acid and oleanolic acid were found identified as the constituents responsible for this synergistic activity.138
Similarly, promiscuous efflux pumps promote resistance by extruding a wide array of compounds from bacterial cells.139,140 In their hallmark paper, Stermitz et al.18 described the presence of an inhibitor of the norA efflux pump, 5′-methoxyhydnocarpin, in Berberis species that potentiated the activity of the efflux pump substrate berberine. More recently, the berberine-containing plant Hydrastis canadensis L. (Ranunculaceae) was found to possess synergistic norA efflux pump inhibitory activity.141 Many of these synergists have been characterized in subsequent publications.17,22,142
Despite the historical effectiveness of bioassay-guided fractionation,150 loss of activity during fractionation is very common.145,148 Additionally, because structural information is not used to guide separations, this approach may result in the repeated isolation of previously described molecules.148 To avoid re-isolation of known molecules, preliminary structural assessment steps to identify and discard samples containing known active constituents can be taken.148,151–153 This process, termed “dereplication,” enables prioritization of samples likely to contain new biologically important entities, facilitating efficient use of resources for compound discovery.148,151–153 Dereplication is often achieved by comparing the spectral patterns of mixture constituents through mass spectrometry,151,154–156 NMR,157 or UV spectroscopy,151 and searching for known compounds with matching spectral fingerprints in a dereplication database. Recently, the Global Natural Product Social Molecular Networking (GNPS) platform has been developed that enables spectral annotation and identification of related compounds using MS/MS molecular networking.148,153,158 In addition, GNPS provides researchers the ability to share raw MS/MS spectra online, enabling crowdsourced spectra annotation and knowledge sharing between laboratories around the world.153
Fig. 5 Synergy-directed fractionation workflow. Reproduced with permission from Junio et al. 2011.17 |
In mass spectrometry-based biochemometrics studies, the number of variables (ions detected) tends to greatly outnumber the number of samples analyzed (i.e. extracts or simplified fractions), posing a problem for many multiple regression models.163 Partial least-squares (PLS) analysis, however, due to its combination of principal component analysis (PCA) and multiple regression analysis, is less affected by this mismatch and is the most popular tool for modelling biochemometric data.163 The resulting PLS models, however, are often incredibly complex and difficult to decipher. Numerous data visualization tools have been developed to extract meaningful information from PLS datasets.146,163–165
One commonly used tool is the S-plot, in which correlation and covariance of variables with a given biological activity are plotted. In a recent study, S-plots were utilized to identify differences in metabolite profiles (detected using UPLC-QTOF-MS) of Garcinia oblongifolia Champ. Ex Benth. (Clusiaceae) leaves, branches, and fruits and to correlate those differences to differences in biological activity.162 Using this approach, 12 marker compounds were identified, primarily xanthones, that were likely responsible for the enhanced antioxidant and cytotoxic properties of the branch extract over other plant parts.162 In another study, S-plots were generated from bioactivity and chemical profiles of the fungus Ganoderma sinense to identify potential anti-tumor agents. This approach successfully identified five known cytotoxic compounds with significant antitumor potential.161 A recent study compared the use of S-plot analysis with an additional data visualization tool, the selectivity ratio, to identify antimicrobial constituents from the fungal organisms Alternaria and Pyrenochaeta sp.146 In this study, both S-plot and selectivity ratio analyses identified macrosphelide A as the dominant bioactive constituent from Pyrenochaeta sp. However, when attempting to identify bioactive compounds from Alternaria sp., the selectivity ratio outperformed the S-plot in its ability to identify altersetin, a low abundance antimicrobial constituent, without being confounded by highly abundant (and only weakly active) constituents in the mixture.146 In a follow up study, an inactive mixture was spiked with known antimicrobial compounds to identify the impact of data acquisition and data processing parameters on biochemometric analysis using the selectivity ratio plot.159 This study found that data transformation, contaminant filtering, and model simplification tools had major impacts on the selectivity ratio models, emphasizing the importance of proper data processing approaches for extracting reliable information from biochemometric datasets.159 In all selectivity ratio studies applied to identify bioactive natural products,146,159,160 bioactive mixture constituents were identified early in the fractionation process, enabling chromatographic isolation efforts to be tailored to mixture constituents that were most likely to possess bioactivity.
These numerous examples illustrate the efficacy of biochemometrics for distinguishing between active and inactive chemical entities in complex mixtures. However, these approaches do not provide structural information about putative unknown active constituents, hindering the ability to truly optimize isolation efforts. To address this gap, a recent study utilized a combination of selectivity ratio analysis and GNPS molecular networking to identify putative active constituents from the botanical medicine Angelica keiskei (Miq.) Koidz. (Apiaceae) and the molecular families to which they belonged.160 Using this approach, a subset of chalcone analogs were targeted for isolation, yielding two known antimicrobial constituents and an additional, low-abundance compound not previously known to possess antimicrobial activity.160 This concept was streamlined into a process called “bioactive molecular networking,” in which bioactivity predictions are directly visualized in molecular networks themselves, where the size of individual nodes correspond to the predicted bioactivity score for each ion (Fig. 6).148 By including both MS/MS fragmentation data and peak area data in the production of molecular networks, bioactive molecular networking enables dereplication, compound annotation, and identification of putative active compounds in one step.148
Fig. 6 Bioactive molecular networking in which nodes connected in a network represent structurally related compounds based on MS/MS fragmentation patterns, and the size of nodes represents the correlation of compound peak areas with biological activity of interest. Figure is reprinted with permission from Nothias et al. 2018.148 |
An additional approach, Compound Activity Mapping, was developed by the Linington laboratory that utilizes image-based cytological screening data and high-resolution mass spectrometry-based metabolomics data to predict both the identities and biological functions of putative bioactive constituents early in the fractionation workflow.147 Using Compound Activity Mapping, biological and chemical datasets are integrated to identify putative bioactive constituents that show consistent positive correlation with phenotypes of interest (Fig. 7).147 The data are presented as a network display, enabling identification and prioritization of lead compounds, even those of low abundance, that likely contribute to a specific biological activity. The utility of this approach was demonstrated through the investigation of 234 extracts of actinobacterial origin.147 Using Compound Activity Mapping, biological and chemical datasets from these samples were combined to identify 13 clusters of bioactive fractions containing 11 known molecular families and four new compounds. Subsequent isolation efforts targeted towards these new compounds revealed the presence of a new natural product family, the quinocinnolinomycins, which were predicted to elicit a cytotoxic response through the induction of endoplasmic reticulum stress.147
Fig. 7 Compound activity mapping workflow. (A) Network analysis of the full chemical space of the tested actinobacterial extracts. Light blue nodes represent extracts connected to all m/z features (red), illustrating the immense chemical complexity of the extract library. (B) Activity histograms and cluster scores for all m/z features. (C) Compound activity map, displaying only extracts and m/z features predicted to be responsible for consistent phenotypes of interest. (D) Close up of a specific bioactive cluster, belonging to the staurosporine natural product family. This figure is reprinted with permission from Kurita et al. 2015.147 |
To identify synergists and additives in complex botanical mixtures, Britton et al. recently combined biochemometric analysis with synergy-directed fractionation to identify mixture components from Hydrastis canadensis that enhanced the antimicrobial efficacy of berberine through additive or synergistic mechanisms.22 In this study, mass-spectrometry datasets were combined with biological assay data to produce selectivity ratio plots predicting putative additives and synergists (Fig. 8). In these plots, negative selectivity ratios are indicative of biological activity, because growth inhibition data (where smaller values indicate biological activity) were used to guide the models. Unlike other biochemometric studies of its kind,146,160 the biological activity data used in this study did not measure of antimicrobial activity, per se, but was rather a measure of each sample's ability to improve the antimicrobial efficacy of berberine. Using this approach, six flavonoids not previously identified using synergy-directed fractionation approaches alone17 were identified as putative additives or synergists. Of these, one compound, predicted by selectivity ratio models to be the top contributor to activity, was isolated and characterized for the first time and its activity as a synergist confirmed. Notably, this compound possessed no antimicrobial activity on its own and may have been missed using biochemometric analyses guided by antimicrobial data alone.22
Fig. 8 Selectivity ratio plots for first, second, and third stages of fractionation [(A–C), respectively] of the botanical Hydrastis canadensis. Growth inhibition data were used to guide selectivity ratio analysis, so variables with negative selectivity ratio are most likely to possess additive or synergistic activity. Known flavonoids (likely to be synergists) are marked in green, while known alkaloids (likely to be additives) are marked in red. First-stage (A) and second-stage (B) models were not able to identify known compounds as contributing to activity. However, the third-stage model (C) predicted seven flavonoids (1, 2, 3, 5, 6, 8, 29) and three alkaloids (10, 22, 23) to possess additive or synergistic activity. With this approach, a new synergistic flavonoid (29) was identified in H. canadensis, and known flavonoids and alkaloids not previously known to possess additive or synergistic activity were prioritized for future studies. This figure is reprinted with permission from Britton et al. 2017.22 |
Efflux pump inhibition assays, like many other assays used in classical drug discovery approaches, test compounds or mixtures one at a time to identify compounds with promising biological activity. To improve efficiency of these methods, mixtures of drugs can be simultaneously evaluated, but identifying which molecules in these mixtures exert biological effects can be challenging.174 To overcome this limitation, pulsed ultrafiltration mass spectrometry (PUF-MS) was developed, which enables screening of mixtures such as natural products and synthetic combinatorial libraries.174 PUF-MS involves the incubation of small molecule mixtures with a target protein in solution. Those molecules with affinity for the target will bind to the protein, and compounds that are not bound can be washed away using an ultrafiltration membrane.174 This approach, though effective, is slowed by the ultrafiltration step. To improve the speed of screening, a Magnetic Microbead Affinity Selection Screening (MagMASS) protocol was developed, in which the protein target of interest is not free in solution, but rather is bound to magnetic beads.175 To separate compounds with and without affinity for the given target, the receptor-bound fraction can be held in solution using a magnet.175 In a recent study, PUF-MS and MagMASS were compared, and both screening methods were found to reliably identify ligands of a specific molecular target from complex botanical matrices.175 MagMASS showed a 6-fold faster separation of bound and unbound compounds when compared to PUF-MS and is compatible with a 96-well plate format.175 Notably, these methods do not require molecules to bind to a particular active site on the target of interest, and can identify ligands that bind to active or allosteric sites. In this way, the assay could be modified to identify combination effects in which the protein's activity is changed through allosteric activation or inhibition.175 However, given that this approach utilizes protein targets rather than whole cells, the combination effects discovered may not translate to intact biological systems. Additionally, promiscuous inhibitors may cause false positive results using this method, requiring orthogonal approaches to confirm validity of results. Furthermore, these approaches require access to purified material of the protein target of interest. Therefore, while these assays provide target-specific information that can generate valuable hypotheses on mechanisms of action, methods based on PUF-MS and its iterations are not applicable for situations where the target of the active compound is either not known or not available.
The use of DNA and RNA microarrays is another popular tool for probing combination effects within complex mixtures, enabling identification of genes that are up- or down-regulated by natural product extracts alone and in combination. In a recent study, an RNA microarray of neuroglia cells was utilized to compare the number of genes impacted by treatment with Andrographis paniculata (Burm.f.) Nees (Acanthaceae), Eleutherococcus senticosus (Rupr. & Maxim.) Maxim. (Araliaceae), and their fixed combination Kan Jang.177 Results illustrated that A. paniculata and E. senticosus deregulated 211 and 207 genes, respectively, 36 of which were common to cells treated with each extract. Using this information, researchers expected that 382 genes would be deregulated in cells treated with the fixed combination Kan Jang. However, only 250 genes were deregulated in Kan Jang treated cells, 111 of which were unique to the Kan Jang combination, potentially due to synergistic interactions between A. paniculata and E. senticosus. Alternatively, 170 genes were only affected by treatments with A. paniculata or E. senticosus and not by the Kan Jang mixture, possibly due to antagonistic interactions between the plant species when applied in combination.177 Importantly, microarray analyses do not provide infallible evidence that genes induced by treatments are responsible for physiological effects or mechanisms of synergy, but provide a framework for future research.
Because of the material- and time-consuming nature of biological testing, in silico approaches have been developed that enable prediction of activity and mechanism of action without the need for direct biological testing. Existing experimental activity data can be used to mine ligand–target relationships and reveal potential biological activities of diverse molecules.178 Key to the success of this approach for identifying putative mechanisms of action is the availability of compound databases that will facilitate sharing of data and innovation in drug discovery research with both single-target and multi-target approaches.178 Similarly, computational approaches including molecular docking, pharmacophore modelling, and similarity searching can be used as so-called “virtual screening” techniques to identify candidate compounds for follow-up testing.178 Of course, these techniques are subject to error and may not provide accurate representation of the biological system in question, particularly if the model datasets are based on incorrect literature-based annotations of compound activities and/or incomplete understanding of molecular processes of disease.
A systems biology-based approach, network pharmacology, predicts the complex interactions between small molecules and proteins in a biological system, and shows potential as a way to evaluate pharmacological effects of natural product mixtures.178 Unlike the classic “silver bullet” approach where single-target mechanisms are identified for single drugs, network pharmacology focuses on multiple constituents with multiple targets. Several studies have successfully utilized network pharmacology to putatively identify active constituents with both known and unknown molecular targets.166,176,178,179 Networks can be built using existing literature data, computationally-derived data, or experimental data. The predictive accuracy of the resulting networks relies on the completeness of databases, the robustness of the computational models, the understanding of the underlying mechanisms of disease, and/or the chosen biological assay.178
Recently, a broad-scale approach was developed in which functional signature ontology (FUSION) maps are utilized to classify putative mechanisms of action of natural products.180 With this method, cellular responses to natural product treatment can be tracked by measuring gene expression of a small, representative subset of genes that provide insight into the physiological state of the cell. The resulting data can then be combined into FUSION maps capable of linking putative bioactive molecules to the proteins and biological pathways that they target in cells.180 This approach has been successfully utilized to link natural products to their mechanisms of action180 and to identify a marine-derived natural product that inhibits AMPK kinase activity in colon tumor cells.181
A similar approach, the Connectivity Map, or CMap, was developed in which genes, drugs, and disease states are connected based on the gene expression fingerprints that they share.182 Originally produced using 164 drugs and mRNA expression profiling, the CMap has since been expanded more than 1000-fold, and now contains over 1.3 million publicly available profiles. This scale-up was achieved using a high-throughput, reduced representation in which only 1000 landmarks are assessed rather than the full transcriptome. This approach, termed L1000, is sufficient to recover 81% of the information contained in the full transcriptome. The L1000 approach offers advantages over popular approaches such as gene expression microarrays and RNA sequencing because of its low cost and hybridization-based nature, making detection of low-abundant transcripts possible without the need for deep sequencing.182 Preliminary testing has illustrated the potential of the expanded L1000 CMap to determine the mechanisms of action of small molecules based on the similarities of their genetic perturbations to those of compounds with known activities. This approach can also be utilized to identify potential off-target effects of a drug or drug combinations.182
During a pilot study, the L1000 CMap was successfully utilized to recover known mechanisms of action from 63% of existing drugs under analysis, to identify the mechanism of action of a previously uncharacterized compound, and to identify compounds with a particular activity of interest. Importantly, the L1000 CMap is not infallible, and 37% of small molecules with known mechanisms of action were not linked to their expected targets during this study. The authors suggest six reasons for this failure: (1) incomplete inhibition of the target by the compound, (2) off target effects, (3) incomplete information in the L1000 data, (4) incorrect data in the literature, (5) biological differences between complete loss of function and loss of a specific protein function, and (6) existence of previously unrecognized connections with stronger connections than expected ones.182 Despite these limitations, the preliminary results of this study emphasize the potential of the L1000 CMap as a launching point for both target- and ligand-based drug discovery.182 Although they have not been explicitly applied to identify mechanisms of synergy or antagonism, the utilization of FUSION maps and the L1000 CMap platform may represent useful tools to enable identification of genes and pathways impacted by a synergistic/antagonistic combination, providing insight into potential mechanisms of action in complex natural product mixtures.
Metabolomics and biochemometric approaches are promising tools for studying synergy, and have just begun to be applied to identifying constituents that participate in combination effects.22 While useful, biochemometric models are subject to limitations based on the biological assays and reference models used to define biological activity. Similarly, the linear regression models used to predict active constituents are inherently limited given that true linear relationships rarely exist, particularly when assessing complex mixtures with numerous unknown combination effects. The application of statistical tools capable of identifying non-linear relationships will be helpful for future research initiatives. In addition, untargeted approaches to identify molecular targets of synergy and unravel synergistic (or antagonistic) mechanisms of action have just begun to be explored, and continued studies on this topic are of the utmost importance. Advancements in Big Data approaches show great promise for identifying active mixture constituents, characterizing the nature of their interactions, and elucidating their potential mechanisms of action. Integrated technologies capable of completing all of these tasks simultaneously remain to be developed. The production of such integrated techniques will become increasingly important in our continued pursuit to understand the biological activities of complex mixtures.
This journal is © The Royal Society of Chemistry 2019 |