Skip to main content
PLOS ONE logoLink to PLOS ONE
. 2020 Jul 9;15(7):e0235929. doi: 10.1371/journal.pone.0235929

Guided screen for synergistic three-drug combinations

Melike Cokol-Cakmak 1, Selim Cetiner 1, Nurdan Erdem 1, Feray Bakan 2, Murat Cokol 1,¤,*
Editor: Luis Eduardo M Quintas3
PMCID: PMC7347197  PMID: 32645104

Abstract

Combinations of three or more drugs are routinely used in various medical fields such as clinical oncology and infectious diseases to prevent resistance or to achieve synergistic therapeutic benefits. The very large number of possible high-order drug combinations presents a formidable challenge for discovering synergistic drug combinations. Here, we establish a guided screen to discover synergistic three-drug combinations. Using traditional checkerboard and recently developed diagonal methods, we experimentally measured all pairwise interactions among eight compounds in Erwinia amylovora, the causative agent of fire blight. Showing that synergy measurements of these two methods agree, we predicted synergy/antagonism scores for all possible three-drug combinations by averaging the synergy scores of pairwise interactions. We validated these predictions by experimentally measuring 35 three-drug interactions. Therefore, our guided screen for discovering three-drug synergies is (i) experimental screen of all pairwise interactions using diagonal method, (ii) averaging pairwise scores among components to predict three-drug interaction scores, (iii) experimental testing of top predictions. In our study, this strategy resulted in a five-fold reduction in screen size to find the most synergistic three-drug combinations.

Introduction

The treatment of many pathogens and tumors involve drug cocktails of three or more combinations. Such combinations are chosen primarily due to their non-overlapping resistance mechanisms [1,2] and the drug interaction (synergy/antagonism) phenotype [36]. Resistance is informed primarily by the mechanism of action of individual drugs. However, a drug combination’s interaction phenotype is determined by experimentation and the astronomically large space of possible high-order combinations precludes comprehensive screens [7,8].

Pairwise drug interactions have been traditionally measured by checkerboard assays, where two drugs are combined in a 2D matrix [9,10]. While still the gold standard for discovering drug synergy, this method is both time and resource intensive. A miniaturized checkerboard reduces the experimental cost of the pairwise interaction measurement by reducing the number of tested combinations [11]. However, even with the more efficient checkerboard assays, the testing of high-order interactions is impractical. An alternative way of measuring drug interactions is the diagonal assay, which samples the most informative regions of the checkerboard assay, thereby reducing the resource use [12,13]. Moreover, the diagonal assay can also be used to efficiently measure high-order interactions [14].

Even with the development of efficient interaction assays, a comprehensive screen for all high-order combinations for drugs targeting a given disease remains elusive. Among 10 drugs, there are only 45 pairwise combinations. However, there are 120 three-drug and 252 five-drug combinations. For any set with more than 10 drugs, the experimental screen for all possible high-order combinations quickly becomes unfeasible. However, several recent studies showed that pairwise interactions can provide reliable estimates for high-order interactions [1417]. Therefore, comprehensive experimental testing of only pairwise interactions, predicting high-order interactions and experimentally validating the most promising predictions may be an efficient method to discover high-order synergies.

When three drugs A, B, and C are combined, the observed (nominal) interaction has four components: Three pairwise interactions (A+B, A+C, B+C) and the interaction resulting specifically from the combination of all three components (A+B+C). This specific interaction resulting only from the high-order combination has been referred to as “emergent interaction [13, 18].” These four components can be expressed as: nominal interaction = emergent interaction + mean (three pairwise interactions). Moreover [13], multiple studies have shown that emergent interactions are rare [6,14]. Therefore, simple mean of pairwise interactions may provide a reliable estimate for nominal three-drug interaction scores.

Here, we tested this approach by first establishing that diagonal assays and checkerboard assays are in agreement for a large set of pairwise interactions among eight compounds. Using the pairwise interaction scores, we predicted three-drug interaction scores, which we experimentally confirmed. Our results suggest that a search for high-order interactions may require only diagonal assays, and checkerboard assays can be used for validation purposes. Moreover, our results show that the strongest high-order synergies are concentrated among the top predictions, which greatly reduces the experimental search space for high-order drug synergies.

We used Erwinia amylovora, which causes fire blight, as the biological agent in our study. This bacterial pathogen costs to the agricultural economy more than $100 million in the USA [19] per year by destroying young apple and pear trees. We used a set of eight compounds for the screens described in this study. Six of these compounds (copper sulphate, gentamicin, kasugamycin, oxolinic acid, oxytetracycline, and streptomycin) have been previously used against fire blight [2022]. Pentamidine, which has been found to be frequently synergistic in past screens, was also included in this list to increase the likelihood of finding synergistic interactions [23, 24]. Previous studies have suggested a link between detergent compounds and increased synergy [25] in foodborne pathogens. Therefore, we included SDS, a detergent, in our interaction screen. Although the current study focuses on E. amylovora and eight compounds against this pathogen, our guided screen can be used to find synergistic three-drug combinations in other biological and chemical contexts.

Results

Screen of all pairwise combinations among 8 compounds using miniaturized checkerboard assays

In these assays, E. amylovora cells were grown in 4x4 grids where the concentration of each drug is linearly increased along each axis from zero to close to Minimum Inhibitory Concentration (MIC). Growth at each concentration combination was normalized to growth at the no drug condition (see Methods [11]). The concavity of isophenotypic growth contours are analyzed to obtain synergy scores [9, 24]. Under the Loewe additivity model, drug combinations with linear isophenotypic contours are defined as additive (non-interacting). When the isophenotypic contour is concave, relatively less drug is needed to achieve the same effect, defined as synergy. Conversely, convex isophenotypic contours correspond to drug antagonism. Names, abbreviation and top concentration for the 8 drugs used in this study are given in Table 1. Example experimental results for a synergistic pair (oxo+SDS) and an antagonistic pair (cos+oxy) are shown in Fig 1a.

Table 1. Names, abbreviation, top concentration and class of the drugs used in this study.

COMPOUNDS ABBREVIATION MIC (MG/ML) CLASS
COPPER (II) SULFATE cos 0.5 Inorganic compound
GENTAMICIN SULFATE gen 0.003 Aminoglycoside antibiotic
KASUGAMYCIN kas 0.14 Aminoglycoside antibiotic
OXOLINIC ACID oxo 0.00003 Quinolone antibiotic
OXYTETRACYCLINE oxy 0.0002 Tetracycline antibiotic
PENTAMIDINE pen 0.016 Antiprotozoal antibiotic
SODIUM DODECYL SULFATE SDS 0.15 Synthetic organic compound
STREPTOMYCIN SULFATE str 0.0042 Aminoglycoside antibiotic

Fig 1. Measurement of all pairwise interactions among eight drugs in E. amylovora, using checkerboard assays.

Fig 1

(a) In these assays, cells are grown in a 4x4 matrix of the combinations of two drugs. The concentration of each drug linearly increases from 0 to the minimum inhibitory concentration (MIC) in one axis. Here, 16-hour growth curves are shown in white for a synergistic pair (oxo+SDS) and an antagonistic pair (cos+oxy) observed in our screen. Growth is defined as the area under the growth curve (AUC) normalized to the AUC of the no drug condition and is represented by a black (no growth) to red (growth in no drug condition) colorbar. We identified the largest isophenotypic contour that connects the observed phenotypes in single drugs. When this isophenotypic contour is linear, two drugs are additive; concave or convex contours (shown in blue) correspond to synergy (oxo+SDS) or antagonism (cos+oxy), respectively. The three-letter abbreviations for all drugs were given in Table 1. (b) Observed growth in the 4x4 checkerboard assays for all 28 pairwise combinations among eight drugs are shown as heatmaps. In each combination, observed growth in two replicates are shown as two triangles. For each checkerboard assay, the concavity of the largest isophenotypic contour was quantified as α, which is negative, 0 or positive for synergistic, additive or antagonistic pairs, respectively. The inset shows a scatter plot for the replicate α scores which showed strong correlation (Spearman r = 0.73, p-value = 1.5 x 10−5). Green and magenta lines correspond to synergy (α < -1) and antagonism (α > 1) thresholds. SDS exhibited strong synergy with oxo, oxy and kas. Most combinations were antagonistic, in agreement with previous pairwise interaction screens.

We conducted miniaturized checkerboard assays for all 28 pairwise combinations of 8 drugs, in duplicate. Fig 1b shows all the growth measurements for all experiments for both duplicates, which show remarkable agreement. We observe that concave isophenotypic contours (synergy) are rare while convex contours (antagonism) are common, in agreement with numerous studies [8, 23, 24]. The few synergies involve SDS, with pairwise combinations of oxy, kas or oxo showing striking concavity. Cos was antagonistic with all drugs including SDS. We used a previously established score (α) to quantify the concavity of the largest isophenotypic contour observed in each miniaturized checkerboard assay [26]. α is 0 for linear (additive) pairwise combinations; negative or positive for concave (synergistic) or convex (antagonistic) combinations. Fig 1b inset shows the duplicate α scores obtained for all pairs. Two replicate scores significantly correlated (Spearman r = 0.73, p < 0.01), supporting the reliability of the checkerboard screen results.

Screen of all pairwise combinations among 8 compounds using diagonal assays

In these assays, cells are grown in increasing concentrations of individual compounds to determine the dose fractions that result in a given phenotype, such as 50% growth inhibition. Using these values, an expected dose-fraction that would result in the same phenotype for a 1:1 mixture of two compounds is computed under the assumption of additivity among these compounds. To assess synergy, the experimental dose-fraction obtained from a 1:1 mixture of two compounds is compared with the expected dose-fraction. If observed dose-fraction is smaller than, equal to, or larger than the expected dose-fraction, synergy, additivity or antagonism is concluded, respectively (Fig 2a) [12,13]. In Fig 2b, experimental results for a synergistic pair (oxo+SDS) and antagonistic pair (cos+pen) are shown.

Fig 2. Measurement of all pairwise interactions among eight drugs in E. amylovora using diagonal assays.

Fig 2

(a) In these assays, cells are grown in linearly increasing doses of two single drugs (A and B) and an equipotent mixture of these two drugs. Growth is represented by a black (no growth) to red (growth in no drug condition) colorbar. The observed dose that results in 50% growth inhibition (IC50) is indicated by a white circle. The expected IC50 of the combination is indicated by a black triangle. When drugs are mixed, the observed IC50 of the combination may be lower, equal or higher than the expected IC50, corresponding to synergy, additivity or antagonism, respectively. λ score is defined by log2(obs IC50 / exp IC50), hence negative, zero or positive values correspond to synergy, additivity or antagonism, respectively. (b) Examples of observed synergy and antagonism. The observed IC50 of oxo+SDS combination is smaller than the observed IC50s of single compounds and the expectation calculated for the combination, indicating synergy. cos+pen has almost no effect while both single drugs cos and pen have growth inhibition effects, indicating antagonism. The three-letter abbreviations for all drugs were given in Table 1. (c) Summary of diagonal method results for all pairwise interactions. We projected the observed IC50 in single drugs and two-drug combination in each experiment to a 2D matrix and connected these three points to construct an isophenotypic contour. Red and black contours correspond to two replicates. Similar to the checkerboard assay interpretation, concave or convex contours indicate that the drug combination is synergistic or antagonistic, respectively. The inset shows that replicate λ scores strongly correlate (Spearman r = 0.79, P-value = 2.2 x 10−6). Green and magenta lines correspond to synergy (λ < -0.5) and antagonism (λ > 0.5) thresholds.

We conducted diagonal assays for all 28 pairwise combinations of 8 drugs, in duplicate. Fig 2c shows a visual representation of the data obtained from these assays, where the observed dose-fraction for all single and pairwise combinations are projected on a 2D matrix. This representation allows to approximate the concavity of the isophenotypic contour as determined by the efficient sampling of the diagonal assay. Similar to checkerboard assays, concave or convex contours correspond to synergy or antagonism, respectively. cos was antagonistic in all experiments. In agreement with the checkerboard assays, antagonism was common. We used a previously established score (λ) to quantify the synergy for each experiment, where λ = log2(observed dose-fraction / expected dose-fraction). In this setting, a score of 0 corresponds to additivity. Negative and positive scores correspond to synergy or antagonism, respectively. Fig 2c inset shows that the duplicate λ scores for all pairs significantly correlated (Spearman r = 0.79, p < 0.01), supporting the reliability of the diagonal assay results. Next, we compared the α scores obtained from checkerboard assay screen with the λ scores obtained from diagonal assay screen. We used the mean of α or λ values for each pair and found that α and λ scores significantly correlated (Spearman r = 0.53, p < 0.01), supporting the use of diagonal assays as an efficient proxy for checkerboard assays in measuring drug interactions.

Prediction and testing of three-drug interactions using pairwise interaction scores

For all three-drug combinations of these 8 antibiotics, we generated predictions by using the arithmetic mean of interaction scores for all 3 pairwise combinations among 3 drugs, as introduced above. For combination A+B+C, the mean (AB, AC, BC) provides the expectation for A+B+C if there is no additional synergy/antagonism associated with the three-drug combination of these drugs. Since we planned to test our predictions using the diagonal method, we only used pairwise λ scores (λ2) for our predictions. The experimental setup and interpretation of three-drug interactions using the diagonal method is similar to the pairwise example with three individual dose responses and 1:1:1 mixture vs. two dose responses and 1:1 mixture (Fig 3a). Fig 3b shows examples for three-drug synergy (oxo+oxy+SDS) and three-drug antagonism (oxo+oxy+str).

Fig 3. Measurement of all three-drug drug interactions among seven drugs in E. amylovora, using diagonal assays.

Fig 3

(a) Similar to the diagonal assay for pairwise interactions, cells are grown in linearly increasing doses of three single drugs and one equipotent mixture of these three drugs. An expected IC50 is calculated using the IC50 of the individual drugs. If observed IC50 is smaller or larger than expected IC50, synergy or antagonism is concluded, respectively. (b) All three-way interactions are represented as 3D heatmap. Green, white or magenta boxes correspond to synergistic, additive or antagonistic three-drug drug combinations. On the right, the 3D heatmap is shown as layers to assist visualization.

To test our predictions, we experimentally tested all three-drug combinations among seven drugs (35 combinations). cos was not included in this screen as all of its interactions were predicted to be antagonistic, hence decreasing the screen size from 56 to 35. The three-drug interaction score (λ3) measured for each combination is shown in Fig 3c. As in the pairwise screen, we found that the most synergistic three-drug combinations involved SDS.

Comparison of predicted and empirical scores

Fig 4a shows a scatter plot for predicted scores mean (λ2) and experimental scores (λ3) for all 35 combinations used in the screen. mean(λ2) and λ3 significantly correlated (Spearman r = 0.5, p < 0.01), indicating that pairwise interaction scores provide reliable estimations for three-drug interactions. Therefore, screening only a subset of three-drug combinations guided by pairwise interactions emerges as an efficient platform to discover three-drug synergies. For each three-drug combination depicted in Fig 4a, their percentile in mean(λ2) ranking is indicated. For example, the combination that is predicted to have the lowest λ3 is at ~3 percentile, since there are 35 combinations considered. Of the 35 three-drug combinations, two were found to have the strongest synergies (kas+oxo+SDS) and (oxo+oxy+SDS), and were in the top 20th percentile of predicted synergies. These results show that testing only the top seven combinations predicted by our method would have been sufficient to identify the most synergistic three-drug combinations in the entire three-drug combination space, which corresponds to a five-fold increase in the efficiency of synergy screening.

Fig 4. Prediction of three-drug interactions using the average of pairwise interaction scores.

Fig 4

(a) Each circle represents a tested three-drug interaction (A+B+C). x-axis shows the mean of pairwise interaction scores (A+B, A+C, B+C) λ2 and y-axis shows the empirically obtained three-drug interaction score λ3. Mean(λ2) and λ3 significantly correlated, indicating that three-drug interactions can be predicted using only pairwise interaction scores (Spearman r = 0.50, P-value = 2.1 x 10−3). kas+oxo+SDS and oxo+oxy+SDS were the strongest three-drug synergies. On each three-drug interaction, the percentile among mean(λ2) scores is indicated. kas+oxo+SDS and oxo+oxy+SDS are in the top 20 percentile. Green and magenta lines correspond to synergy (λ < -0.5) and antagonism (λ > 0.5) thresholds. (b) Three-drug interaction scores λ3 and emergent interaction scores (λ3—mean(λ2)) strongly correlate, indicating that emergent interactions contribute to three-drug interactions (Spearman r = 0.75, P-value = 1.7 x 10−7). (c) Mean(λ2) and emergent interaction scores had no significant correlation.

To test specificity, we used shuffled pairwise interaction data to predict 3-drug interaction scores. In 100,000 simulations, predictions derived from shuffled pairwise data sets showed a correlation equal to or more than real data in 3% of the cases. In addition, we used real pairwise interaction data to predict shuffled 3-drug interaction scores. In 100,000 simulations, predictions showed a correlation equal or more than real data in 1.2% of the cases. The corresponding p-values 0.03 and 0.01 attest to the specificity of our predictions.

The difference between mean (λ2) and λ3 scores has been named as emergent synergy, since this interaction emerges only as a result of the three-drug combination [13, 27]. For each three-drug combination, we computed the emergent λ3 score by = λ3 –mean(λ2). Fig 4b shows that emergent λ3 scores highly correlate with λ3 (Spearman r = 0.75, p < 0.01). Therefore, emergent interactions play an important role for high-order interactions. Emergent interactions do not significantly correlate with mean(λ2). In other words, combinations of synergistic pairs do not result in even more synergistic three-drug interactions. Importantly, the distribution of emergent interactions is skewed towards positive values, therefore three-drug interactions are overall more antagonistic than what is expected from pairwise interactions.

Discussion

A comprehensive experimental screen for high-order synergies is currently unfeasible, even for relatively small sets of drugs. For example, when 20 drugs are considered, there are 1140 three-drug combinations. Recent studies have shown that pairwise interaction scores provide reliable estimates for three-drug interactions. There are 190 pairwise combinations among 20 drugs. For n drugs, there are (n choose 3) possible three-drug combinations and (n choose 2) possible 2-way combinations. (n choose 3) is (n-2)/3 times larger than (n choose k). For n = 20, there are 6 times more three-drug interactions than pairwise interactions. Therefore, the experimental measurement of only pairwise combinations, to prioritize synergistic three-drug candidates is a strategy for efficiently discovering three-drug synergies. Here, we formalized this approach by (i) measuring all pairwise interactions by checkerboard assay, (ii) measuring all pairwise interactions by diagonal assay, (iii) establishing that diagonal method measures the same phenomenon as the traditional checkerboard assay, (iv) predicting three-drug synergy scores using pairwise diagonal method scores, and (v) testing the three-drug predictions using the diagonal method. Since we have already established the correlation between checkerboard assay and diagonal assay, further iterations of this search strategy may exclude checkerboard assays altogether to discover synergistic three-drug combinations with minimal cost.

A comparison of the checkerboard and diagonal assays strongly argues that the diagonal assay should be the first line of experimentation for synergy screens. Checkerboard assays may then be used to evaluate a combination of interest for validation or discovery of asymmetric interactions. For pairwise interactions, diagonal and checkerboard assays use drug concentration gradients in 1D and 2D, respectively. The experimental setup of a 2D gradient is more difficult than a 1D gradient. For data interpretation, checkerboard assays use isophenotypic contour analysis, which is difficult to perform. Interpretation of diagonal assay is a simple division of observed and expected dose-fractions. Whereas checkerboard assays use more resources than diagonal assays for similar information, diagonal assays are more sensitive than checkerboard assays for equivalent resource cost. In our miniaturized checkerboard assays, we used 4 gradients of each drug and 4x4 = 16 microplate wells per combination. In our diagonal assays, we used 8 gradients of each drug (and combination) and used 8x3 = 24 microplate wells per combination. Despite only 33% increase in experimental cost, the diagonal assay has 2X the resolution of the miniaturized checkerboard assay. This is represented by the stronger correlation among replicates for the diagonal assay (0.79), as compared with the replicate correlation of the mini-checkerboard assay (0.73). For high-order combinations, checkerboard assay quickly becomes prohibitively expensive, necessitating the use of diagonal method. A miniaturized 3D checkercube assay would have 4x4x4 = 64 wells. However, measurement of a three-drug interaction with the diagonal method using an 8-dose gradient only takes 4x8 = 32 wells. Even for three-drug interactions, the diagonal method is both cheaper and more sensitive than mini-checkerboard assays.

The observation that pairwise interaction scores provide reliable estimates for three-drug interactions can be formally expressed by the disentanglement of each factor that contributes to the three-drug interaction. When three drugs A, B, and C are combined, all three pairwise interactions (A+B, A+C, B+C) and one three-drug emergent interaction (A+B+C) occur and the observed interaction is a sum of all these interactions, which can be factorized as: λ3 = mean(λ2) + emergent λ3. Our study is in agreement with previous studies that show that mean(λ2) is not correlated with emergent λ3 [13]. Currently, emergent synergy is computationally unpredictable and experimentally more expensive than measuring the combination’s nominal synergy. Importantly, emergent λ3 is distributed around 0 with a tendency for positive values. Therefore, in the absence of emergent λ3 information, most three-drug combinations will be expected to be slightly more antagonistic than the mean of pairwise interactions.

We importantly note that we have thus far only considered synergy/antagonism of the efficacy of a drug combination. Drugs and their combinations may have side effects such as toxicity, and a combination may have synergistic/antagonistic toxicity. Therefore, a good combination treatment would have synergistic efficacy while not having synergistic toxicity [28, 29, 30, 31]. In a recent study we showed that combinations that have synergistic activity for both desired and undesired phenotypes, cautioning against the side effects of synergistic combinations [32]. A good rule-of-thumb while designing combinations is choosing synergistic combinations with non-overlapping toxicity. This design element is similar to choosing combinations with non-overlapping resistance mechanisms [1, 2, 8]. Since both heuristics are informed only by experimentation on single drugs, their solution does not require experimental screens of large number of combinations. Therefore, the guided screen presented here may be extended by the consideration of non-overlapping toxicity and resistance at minimal cost.

While our study is aimed to describe a general method for finding three-drug synergies for any pathogen or tumor, the results we have obtained for E. amylovora are also noteworthy. As in numerous previous screens, we have found that antagonism is common, and synergy is very rare [8, 23, 24]. This attests to the need for prudent design of synergistic drug combinations. The only synergies we identified in our pairwise or three-drug screens involved SDS, which we have included for its synergy inducing properties. Combining antibiotics with soap, of course, is not a viable option for systemic infections in humans. However, SDS is recognized as safe for food use by FDA and is used as a control agent for a fungal pathogen to reduce the white rust of spinach [33]. For this reason, use of SDS+antibiotic combinations in the management of plant infections such as fire blight might indeed be practical and as we have seen in our study, may result in synergistic therapeutic benefits to the plant. A next step may include application of combining SDS with other antibiotics in greenhouse and field experiments.

Materials and methods

Experimental materials and methods

Copper (II) sulfate (Lot # BCBP2427V), gentamicin sulfate (Lot # SLBK1443V), kasugamycin (Lot # BCBS4283V), oxolinic acid (Lot # BCBH6650V), oxytetracycline hydrochloride (Lot # BCBG9599V), pentamidine isethionate salt (Lot # 117K3731V) and streptomycin sulfate (Lot # 081M13801V) were purchased from Sigma. Sodium dodecyl sulfate (Product No: M82553115) was obtained from Molekula. All drugs were dissolved in water from Millipore MilliQ ultrapure water system and kept at -20°C. All experiments were conducted with Erwinia amylovora (Burill) ATCC 49946. Cells were grown in LB at 26°C for 16 hours and diluted to OD600: 0.01. Optical densities of the cells were measured using a plate reader (Tecan Microplate Reader). This microplate reader keeps the temperature at 26°C and provides air flow by shaking the plates in every 15 minutes. MATLAB code was used to obtain the growth curves from the numerical data.

Determination of the Minimum inhibitory Concentration (MIC)

The MIC of eight drugs against E. amylovora was determined by using a cell growth assay in 96- well culture plates using LB growth media, with logarithmic, and then linear dilution of drugs. An automated plate reader (Tecan Microplate Reader) was used to quantitatively measure the turbidity in all wells, and the concentration at which there is no increase in turbidity (no cell growth) was identified as the MIC for that drug. The MIC determined by this method corresponds to the concentration at which the drug is bacteriostatic.

4 × 4 checkerboard assay

After determining the individual MIC of each drugs, pairwise interactions were evaluated according to the method of “Miniaturized Checkerboard Assays to Measure Antibiotic Interactions” [11]. For this, the concentration of each individual drug was linearly increased on each axis ranging from zero to MIC in a 4x4 matrix. All assays were performed in duplicate. OD600 readings was measured in a TECAN Infinite F200 microplate reader every 15 min at 26°C for 16 hours. The α-score for each 4x4 assay were quantified using the isophenotypic growth contour method described [23]. The results were interpreted as synergy (α < -1) and antagonism (α > 1) thresholds. If the growth contour was linear, concavity or convexity, interactions were classified as additive, synergistic or antagonistic, respectively.

Diagonal assays

All pairwise interactions were performed according to the method by “Diagonal Method to Measure Synergy Among Any Number of Drugs” for establishing that diagonal method measures the same phenomenon as the traditional checkerboard assay [12]. This method requires two single and one pairwise drug dose-response, in total. For this, concentration of each individual drugs and an equipotent mixture of these drugs (1:1 v/v) were linearly increased in one axis into 96 well plate. E. amylovora were grown in LB media in the presence of combinations of eight drugs. OD600 measurements using a TECAN microplate reader were done every 15 min at 26°C for 16 hours. Dose-response curves were generated using MATLAB. The expected dose of the two-drug combination was calculated as the intersection of this two-drug-dose-response in the dose combination space with the straight line defined by the IC50 values of each single drug dose-response [34]. The observed dose of the combination is the interpolated IC50 of the two-drug dose-response. λ was calculated using the formula: λ2 = log2 (obs IC50 / exp IC50). The results were interpreted as synergy (λ2 < -0.5) and antagonism (λ2 > 0.5) thresholds.

The synergy scores obtained in pairwise interaction was a guidance in discovery of three-way combinations. For testing the three-drug predictions using diagonal method, the concentration of each drugs and equipotent mixture of these drugs (1:1:1 v/v) were linearly increased in one axis into 96 well plate. Bacteria were treated with the combinations of eight drugs. λ was calculated using the formula: λ3 = log2 (obs IC50 / exp IC50). The results were interpreted as synergy (λ3 < -0.5) and antagonism (λ3 > 0.5) thresholds.

All pairwise interaction experiments (checkerboard and diagonal testing) were done in two biological replicates. The agreement between checkerboard replicates (r = 0.73, p < 0.01) or diagonal replicates (r = 0.83, p < 0.01) are given in Figs 1 or 2, respectively. Our 3-drug predictions were based on diagonal scores, which were significantly but not perfectly correlated. To test predictions for 3-drugs, we calculated the correlation between predictions and empirical interaction scores. Importantly, the correlation between predictions and experiments cannot be higher than the correlation among experiments. Therefore, the highest correlation we may expect from prediction-experiment comparison is 0.83 (correlation among replicates of pairwise diagonal experiments). The correlation of 0.5 between prediction-experiment was able to capture 0.5/0.83*100 = 60% of the predictable correlation despite the underlying noise in the data.

Acknowledgments

The authors thank Elif Ozaltan and Ece Bicak for their assistance in experiments, and Zohar B. Weinstein and Fatih Ozbay for their critical reading on the manuscript.

Data Availability

Experimental data obtained during this study and calculated interaction scores are available from: https://doi.org/10.6084/m9.figshare.12043626.

Funding Statement

This work was supported by FP7 Marie Curie IRG (268440) and Scientific and Technological Research Council of Turkey (110S209) Grants to MC. The specific roles of these authors are articulated in the ‘author contributions’ section. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. No additional external funding was received for this study.

References

  • 1.Palmer AC, Chidley C, Sorger PK. A curative combination cancer therapy achieves high fractional cell killing through low cross-resistance and drug additivity. eLife. 2019. November 19;8:e50036 10.7554/eLife.50036 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Law LW. Effects of combinations of antileukemic agents on an acute lymphocytic leukemia of mice. Cancer research. 1952. December 1;12(12):871–8. [PubMed] [Google Scholar]
  • 3.Zimmermann GR, Lehar J, Keith CT. Multi-target therapeutics: when the whole is greater than the sum of the parts. Drug discovery today. 2007. January 1;12(1–2):34–42. 10.1016/j.drudis.2006.11.008 [DOI] [PubMed] [Google Scholar]
  • 4.Berenbaum MC. What is synergy. Pharmacol Review. 1989;41:93–141. [PubMed] [Google Scholar]
  • 5.Cokol M. Drugs and their interactions. Current drug discovery technologies. 2013. June 1;10(2):106–13. 10.2174/1570163811310020003 [DOI] [PubMed] [Google Scholar]
  • 6.Meyer CT, Wooten DJ, Lopez CF, Quaranta V. Charting the Fragmented Landscape of Drug Synergy. Trends in Pharmacological Sciences. 2020. February 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Wood KB. Pairwise interactions and the battle against combinatorics in multidrug therapies. Proceedings of the National Academy of Sciences. 2016. September 13;113(37):10231–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Yilancioglu K, Cokol M. Design of high-order antibiotic combinations against M. tuberculosis by ranking and exclusion. Scientific reports. 2019. August 15;9(1):1–1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Greco WR. The search for synergy: a critical review from a response surface perspective. Pharmacol Rev. 1995;47:331–85. [PubMed] [Google Scholar]
  • 10.Lehár J, Zimmermann GR, Krueger AS, Molnar RA, Ledell JT, Heilbut AM, et al. Chemical combination effects predict connectivity in biological systems. Molecular systems biology. 2007. January 1;3(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Cokol-Cakmak M, Cokol M. Miniaturized Checkerboard Assays to Measure Antibiotic Interactions In Bioinformatics and Drug Discovery 2019. (pp. 3–9). Humana Press, New York, NY. [DOI] [PubMed] [Google Scholar]
  • 12.Cokol-Cakmak M, Bakan F, Cetiner S, Cokol M. Diagonal Method to Measure Synergy Among Any Number of Drugs. JoVE (Journal of Visualized Experiments). 2018. June 21(136):e57713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Cokol M, Kuru N, Bicak E, Larkins-Ford J, Aldridge BB. Efficient measurement and factorization of high-order drug interactions in Mycobacterium tuberculosis. Science advances. 2017. October 1;3(10):e1701881 10.1126/sciadv.1701881 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Katzir I, Cokol M, Aldridge BB, Alon U. Prediction of ultra-high-order antibiotic combinations based on pairwise interactions. PLoS computational biology. 2019. January 30;15(1):e1006774 10.1371/journal.pcbi.1006774 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Zimmer A, Katzir I, Dekel E, Mayo AE, Alon U. Prediction of multidimensional drug dose responses based on measurements of drug pairs. Proceedings of the National Academy of Sciences. 2016. September 13;113(37):10442–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wood K, Nishida S, Sontag ED, Cluzel P. Mechanism-independent method for predicting response to multidrug combinations in bacteria. Proceedings of the National Academy of Sciences. 2012. July 24;109(30):12254–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wood KB, Wood KC, Nishida S, Cluzel P. Uncovering scaling laws to infer multidrug response of resistant microbes and cancer cells. Cell reports. 2014. March 27;6(6):1073–84. 10.1016/j.celrep.2014.02.007 [DOI] [PubMed] [Google Scholar]
  • 18.Beppler C, Tekin E, Mao Z, White C, McDiarmid C, Vargas E, et al. Uncovering emergent interactions in three-way combinations of stressors. Journal of the Royal Society Interface. 2016. December 31;13(125):20160800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Norelli JL, Jones AL, Aldwinckle HS. Fire blight management in the twenty-first century: using new technologies that enhance host resistance in apple. Plant Disease. 2003. July;87(7):756–65. 10.1094/PDIS.2003.87.7.756 [DOI] [PubMed] [Google Scholar]
  • 20.Sholberg PL, Boule J. Evaluation of antibiotics and plant extracts for control of streptomycin-resistant Erwinia amylovora. InXI International Workshop on Fire Blight 793 2007 Aug 12 (pp. 423–428).
  • 21.Rekanovic E, Milijasevic S, Filajdic N, Gavrilovic V. Efficacy of antibiotics and copper compounds in Erwinia amylovora control in Serbia. InX International Pear Symposium 800 2007 May 22 (pp. 875–878).
  • 22.ZHAO YQ, TIAN YL, WANG LM, GENG GM, ZHAO WJ, HU BS, et al. Fire blight disease, a fast-approaching threat to apple and pear production in China. Journal of Integrative Agriculture. 2019. April 1;18(4):815–20. [Google Scholar]
  • 23.Mason DJ, Stott I, Ashenden S, Weinstein ZB, Karakoc I, Meral S, et al. Prediction of antibiotic interactions using descriptors derived from molecular structure. Journal of medicinal chemistry. 2017. May 11;60(9):3902–12. 10.1021/acs.jmedchem.7b00204 [DOI] [PubMed] [Google Scholar]
  • 24.Cokol M, Chua HN, Tasan M, Mutlu B, Weinstein ZB, Suzuki Y, et al. Systematic exploration of synergistic drug pairs. Molecular systems biology. 2011. January 1;7(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Chen D, Zhao T. Organic acids, detergents, and their combination for inactivation of foodborne pathogens and spoilage microorganisms In Natural and Bio-based Antimicrobials for Food Applications 2018. (pp. 63–85). American Chemical Society. [Google Scholar]
  • 26.Chandrasekaran S, Cokol‐Cakmak M, Sahin N, Yilancioglu K, Kazan H, Collins JJ, et al. Chemogenomics and orthology‐based design of antibiotic combination therapies. Molecular systems biology. 2016. May 1;12(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Tekin E, White C, Kang TM, Singh N, Cruz-Loya M, Damoiseaux R, et al. Prevalence and patterns of higher-order drug interactions in Escherichia coli. npj Systems Biology and Applications. 2018. September 3;4(1):1–0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bulusu KC, Guha R, Mason DJ, Lewis RP, Muratov E, Motamedi YK, et al. Modelling of compound combination effects and applications to efficacy and toxicity: state-of-the-art, challenges and perspectives. Drug discovery today. 2016. February 1;21(2):225–38. 10.1016/j.drudis.2015.09.003 [DOI] [PubMed] [Google Scholar]
  • 29.Chen C, Wang Y, Qian Y, Zhao X, Wang Q. The synergistic toxicity of the multiple chemical mixtures: implications for risk assessment in the terrestrial environment. Environment international. 2015. April 1;77:95–105. 10.1016/j.envint.2015.01.014 [DOI] [PubMed] [Google Scholar]
  • 30.Weinstein ZB, Kuru N, Kiriakov S, Palmer AC, Khalil AS, Clemons PA, et al. Modeling the impact of drug interactions on therapeutic selectivity. Nature communications. 2018. August 27;9(1):1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Cheng F, Kovacs IA, Barabási AL. Network-based prediction of drug combinations. Nature Communications. 2019. March 13;10(1):1197 10.1038/s41467-019-09186-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Lehar J, Krueger AS, Zimmermann GR, Borisy AA. Therapeutic selectivity and the multi-node drug target. Discovery Medicine. 2009. December;8(43):185–90. [PubMed] [Google Scholar]
  • 33.Irish BM, Correll JC, Morelock TE. The effect of synthetic surfactants on disease severity of white rust on spinach. Plant Disease. 2002. July;86(7):791–6. 10.1094/PDIS.2002.86.7.791 [DOI] [PubMed] [Google Scholar]
  • 34.Weinstein ZB, Zaman MH. Quantitative bioassay to identify antimicrobial drugs through drug interaction fingerprint analysis. Scientific reports. 2017. February 16;7:42644 10.1038/srep42644 [DOI] [PMC free article] [PubMed] [Google Scholar]

Decision Letter 0

Luis Eduardo M Quintas

21 May 2020

PONE-D-20-08982

Guided screen for synergistic three-drug combinations

PLOS ONE

Dear Dr. Cokol,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process. More specifically, reviewer #3 made important comments on the statistical analysis.

We would appreciate receiving your revised manuscript by Jul 05 2020 11:59PM. When you are ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter.

To enhance the reproducibility of your results, we recommend that if applicable you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. For instructions see: http://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). This letter should be uploaded as separate file and labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. This file should be uploaded as separate file and labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. This file should be uploaded as separate file and labeled 'Manuscript'.

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out.

We look forward to receiving your revised manuscript.

Kind regards,

Luis Eduardo M Quintas, Ph.D.

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2. Thank you for stating the following in the Financial Disclosure section:

The funders had no role in study design, data collection and analysis, decision to

publish, or preparation of the manuscript. 

We note that one or more of the authors are employed by a commercial company: Axcella Health.

a. Please provide an amended Funding Statement declaring this commercial affiliation, as well as a statement regarding the Role of Funders in your study. If the funding organization did not play a role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript and only provided financial support in the form of authors' salaries and/or research materials, please review your statements relating to the author contributions, and ensure you have specifically and accurately indicated the role(s) that these authors had in your study. You can update author roles in the Author Contributions section of the online submission form.

Please also include the following statement within your amended Funding Statement.

“The funder provided support in the form of salaries for authors [insert relevant initials], but did not have any additional role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript. The specific roles of these authors are articulated in the ‘author contributions’ section.”

If your commercial affiliation did play a role in your study, please state and explain this role within your updated Funding Statement.

b. Please also provide an updated Competing Interests Statement declaring this commercial affiliation along with any other relevant declarations relating to employment, consultancy, patents, products in development, or marketed products, etc. 

Within your Competing Interests Statement, please confirm that this commercial affiliation does not alter your adherence to all PLOS ONE policies on sharing data and materials by including the following statement: "This does not alter our adherence to  PLOS ONE policies on sharing data and materials.” (as detailed online in our guide for authors http://journals.plos.org/plosone/s/competing-interests) . If this adherence statement is not accurate and  there are restrictions on sharing of data and/or materials, please state these. Please note that we cannot proceed with consideration of your article until this information has been declared.

Please include both an updated Funding Statement and Competing Interests Statement in your cover letter. We will change the online submission form on your behalf.

Please know it is PLOS ONE policy for corresponding authors to declare, on behalf of all authors, all potential competing interests for the purposes of transparency. PLOS defines a competing interest as anything that interferes with, or could reasonably be perceived as interfering with, the full and objective presentation, peer review, editorial decision-making, or publication of research or non-research articles submitted to one of the journals. Competing interests can be financial or non-financial, professional, or personal. Competing interests can arise in relationship to an organization or another person. Please follow this link to our website for more details on competing interests: http://journals.plos.org/plosone/s/competing-interests

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: No

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: Methods to better predict higher order drug-drug interactions has multiple applications. Cokol and colleagues have developed several of the methods that have made significant advances in this field. Here, the authors look at 3 drug interactions for 7 treatments for the plant pathogen Erwinia. They identify that only the detergent SDS reliably results in synergistic interactions, with many antagonistic interactions, cautioning against a rationale that ‘multi-drug’ therapy is always superior.

Overall, the study is well-performed and ready for publication.

My only recommendation is that the title is too general, and needs to state that this screen was performed in Erwinia.

Babak Javid

Reviewer #2: The article by Cokol-Cakmak and colleagues addresses an important, old, and for a long time unsolved problem in pharmacology, namely how to choose among the astronomically large number of permutations of multiple drugs that could potentially be used in a combination therapy.

In the domain of antimicrobial and anti-cancer chemotherapy, recent discoveries from multiple groups (including from the senior author) have suggested a possible solution. Specifically, it has been observed in each of bacteria, fungi, and cancer that the most substantial drug interactions occur at the 'pairwise' level, whereas 'higher-order' interactions (among 3 or more drugs) are comparatively minor. The implication of this observation is that the combined potency of cocktails of 3 or more drugs may be predictable from measurement of drug pairs. The current manuscript tests this idea and demonstrates how a synthesis of experiments and predictions makes it possible to systematically search a very large number of potential combinations and find the most strongly synergistic many-drug combinations.

The proposed approach consists of

1) experimentally measure pairwise drug interactions

(using a very experimentally efficient 'diagonal' approach)

2) use pairwise data to predict the effects of high-order combinations

3) experimentally test most promising high-order combinations

This approach is novel and eminently sensible, and I think this article describing and demonstrating this approach will be an important contribution to the field. I am grateful that the approach does not rely on prediction alone, but uses prediction to guide experimental measurements to the most promising options. Theory plays a role but the final result is empirical.

With respect to the potential generality and applicability of the observations, two points are worth noting in review. First, the pharmacological principles that guide this approach have been supported in many different diseases and model systems, and therefore I am satisfied that this proposal on how to practically apply these principles is one that has general interest beyond the particular model system being studied. (The application here to a plant pathogen is outside of my expertise but looks interesting). Second, the use of 'many-drug' combinations is routine in many areas of infectious disease and cancer medicine, so this topic certainly has medical relevance. Any use of novel combinations in humans of course requires phase 1 trials of safety, and combination design for human use should involve considerations of overlapping or non-overlapping toxicities. I think if one were to apply the approach described here to therapies for human use, the approach could take this into account, by narrowing the search space to only one agent per mode of dose-limiting toxicity (i.e. only one agent with dose-limiting hepatotoxicity, only one with dose-limiting cardiotoxicity, etc). So while toxicity is an important consideration in developing human combination therapies it does not override the practicality of the approach here, because these aspects could be considered simultaneously in prediction-guided experiments. Of course this is not relevant to Erwinia amylovora, and I leave it to the authors to decide if they should like to mention these considerations in the discussion.

The most substantial advance of the work is describing and demonstrating the approach to searching for synergistic multidrug combinations. For me the experimental findings in Erwinia amylovora are of secondary significance. The drug interactions observed in this system are of small magnitude making the overall effect of synergy minor. The measurements of pairwise interactions show substantial variation among replicates, although not worse than is typical of high-throughput screens. Although experimental noise in pairwise interactions will propagate into noisy predictions, this concern is overridden by the study ultimately conducting experimental measurements of the high-order combinations of interest. Overall, the strategy described here is interesting, theoretically rigorous, and useful. I expect this article will have general relevance to many efforts to find superior combination therapies for the treatment of pathogens or other diseases.

Reviewer #3: This is an interesting article that may have substantial impact on design of drug combination screens particularly those involving higher order combinations. The article is well written and overall has sufficient clarity. The experiment designs are interesting and does not suffer from any major deficiency to the best of my understanding. There is however need for increased statistical rigor and clarification in certain parts of the paper.

1. The authors claim that their results are “in agreement with numerous studies.” With no justification and in same instances not even a citation. The authors should clarify and detail the nature of the agreement, cite appropriate papers and also justify such agreements. There is also a need for discussion on what such agreements would imply.

2. The data is fairly interesting yet I have not seen in a detailed analysis of biological or technical replicates. The authors need to provide a detailed account of the fit between replicates particularly in the paired combinations and compare the variation/noise in the underlying data to the accuracy of predictions.

3. The authors predicted higher order drug-drug interactions based on the average of paired drug interactions. They explained their strategy as “For all three-drug combinations of these 8 compounds, we generated predictions by using the arithmetic mean of interaction scores for all 3 pairwise combinations among 3 drugs. For combination A+B+C, the mean (AB, AC, BC) provides the expectation for A+B+C if there is no additional synergy/antagonism associated with the three-drug combination of these drugs”. This is an interesting yet extremely simple method. This approach needs better explanation/justification. How did the authors chose this simple approach, what are the examples in the literature, how does their method compare to others? Even If they do not provide a detailed benchmark, a good discussion and scientific justification would strengthen the paper.

4. The prediction power seems to be sufficiently strong based on the analysis in the manuscript. Yet we do not know about the specificity f the predictions. The authors should take a bootstrapping approach to test the predictive power of shuffled data. The predictions from shuffled data will generate a null model of predictions and statistical comparison of their actual predictions to this null model can provide better insight on the value of the predictions.

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

Reviewer #3: No

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files to be viewed.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email us at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2020 Jul 9;15(7):e0235929. doi: 10.1371/journal.pone.0235929.r002

Author response to Decision Letter 0


19 Jun 2020

Reviewer #1: Methods to better predict higher order drug-drug interactions has multiple applications. Cokol and colleagues have developed several of the methods that have made significant advances in this field. Here, the authors look at 3 drug interactions for 7 treatments for the plant pathogen Erwinia. They identify that only the detergent SDS reliably results in synergistic interactions, with many antagonistic interactions, cautioning against a rationale that ‘multi-drug’ therapy is always superior.

Overall, the study is well-performed and ready for publication.

My only recommendation is that the title is too general, and needs to state that this screen was performed in Erwinia.

Babak Javid

We thank the reviewer for his encouraging comments. As all the reviewers noted, the method outlined in our study has applications in multiple fields ranging from antimicrobials, cancer and metabolic diseases. In reviewer 2’s words: “…this proposal on how to practically apply these principles is one that has general interest beyond the particular model system being studied”. We fear that making our title too specific may limit our audience. As we stated in our abstract, the screen is made in Erwinia amylovora, which is just a model for establishing a more general method. For these reasons, we would like to respectfully keep our original title and leave it to the editor’s discretion if a more specific title such as “Guided screen for three-drug combinations in Erwinia amylovora, the causative agent of fire blight” should be used.

Reviewer #2: The article by Cokol-Cakmak and colleagues addresses an important, old, and for a long time unsolved problem in pharmacology, namely how to choose among the astronomically large number of permutations of multiple drugs that could potentially be used in a combination therapy.

In the domain of antimicrobial and anti-cancer chemotherapy, recent discoveries from multiple groups (including from the senior author) have suggested a possible solution. Specifically, it has been observed in each of bacteria, fungi, and cancer that the most substantial drug interactions occur at the 'pairwise' level, whereas 'higher-order' interactions (among 3 or more drugs) are comparatively minor. The implication of this observation is that the combined potency of cocktails of 3 or more drugs may be predictable from measurement of drug pairs. The current manuscript tests this idea and demonstrates how a synthesis of experiments and predictions makes it possible to systematically search a very large number of potential combinations and find the most strongly synergistic many-drug combinations.

The proposed approach consists of

1) experimentally measure pairwise drug interactions

(using a very experimentally efficient 'diagonal' approach)

2) use pairwise data to predict the effects of high-order combinations

3) experimentally test most promising high-order combinations

This approach is novel and eminently sensible, and I think this article describing and demonstrating this approach will be an important contribution to the field. I am grateful that the approach does not rely on prediction alone, but uses prediction to guide experimental measurements to the most promising options. Theory plays a role but the final result is empirical.

With respect to the potential generality and applicability of the observations, two points are worth noting in review. First, the pharmacological principles that guide this approach have been supported in many different diseases and model systems, and therefore I am satisfied that this proposal on how to practically apply these principles is one that has general interest beyond the particular model system being studied. (The application here to a plant pathogen is outside of my expertise but looks interesting). Second, the use of 'many-drug' combinations is routine in many areas of infectious disease and cancer medicine, so this topic certainly has medical relevance. Any use of novel combinations in humans of course requires phase 1 trials of safety, and combination design for human use should involve considerations of overlapping or non-overlapping toxicities. I think if one were to apply the approach described here to therapies for human use, the approach could take this into account, by narrowing the search space to only one agent per mode of dose-limiting toxicity (i.e. only one agent with dose-limiting hepatotoxicity, only one with dose-limiting cardiotoxicity, etc). So while toxicity is an important consideration in developing human combination therapies it does not override the practicality of the approach here, because these aspects could be considered simultaneously in prediction-guided experiments. Of course this is not relevant to Erwinia amylovora, and I leave it to the authors to decide if they should like to mention these considerations in the discussion.

The most substantial advance of the work is describing and demonstrating the approach to searching for synergistic multidrug combinations. For me the experimental findings in Erwinia amylovora are of secondary significance. The drug interactions observed in this system are of small magnitude making the overall effect of synergy minor. The measurements of pairwise interactions show substantial variation among replicates, although not worse than is typical of high-throughput screens. Although experimental noise in pairwise interactions will propagate into noisy predictions, this concern is overridden by the study ultimately conducting experimental measurements of the high-order combinations of interest. Overall, the strategy described here is interesting, theoretically rigorous, and useful. I expect this article will have general relevance to many efforts to find superior combination therapies for the treatment of pathogens or other diseases.

We thank the reviewer for their kind words and their appreciation of our study in the general context of synergy search. We revised our discussion to include the following sentences on overlapping toxicity while designing combinations in humans and a citation to a recent article on the effect of drug interactions on therapeutic selectivity.

“We importantly note that we have thus far only considered synergy/antagonism of the efficacy of a drug combination. Drugs and their combinations may have side effects such as toxicity, and a combination may have synergistic/antagonistic toxicity. Therefore, a good combination treatment would have synergistic efficacy while not having synergistic toxicity. In a recent study we showed that combinations that have synergistic activity for both desired and undesired phenotypes, cautioning against the side effects of synergistic combinations. A good rule-of-thumb while designing combinations is choosing synergistic combinations with non-overlapping toxicity. This design element is similar to choosing combinations with non-overlapping resistance mechanisms. Since both heuristics are informed only by experimentation on single drugs, their solution does not require experimental screens of large number of combinations. Therefore, the guided screen presented here may be extended by the consideration of non-overlapping toxicity and resistance at minimal cost.”

Reviewer #3: This is an interesting article that may have substantial impact on design of drug combination screens particularly those involving higher order combinations. The article is well written and overall has sufficient clarity. The experiment designs are interesting and does not suffer from any major deficiency to the best of my understanding. There is however need for increased statistical rigor and clarification in certain parts of the paper.

1. The authors claim that their results are “in agreement with numerous studies.” With no justification and in same instances not even a citation. The authors should clarify and detail the nature of the agreement, cite appropriate papers and also justify such agreements. There is also a need for discussion on what such agreements would imply.

We apologize for this omission and thank the reviewer for pointing it out. In two instances in the text, we have added citations to support that antagonism is common and synergy is rare.

2. The data is fairly interesting yet I have not seen in a detailed analysis of biological or technical replicates. The authors need to provide a detailed account of the fit between replicates particularly in the paired combinations and compare the variation/noise in the underlying data to the accuracy of predictions.

We thank the reviewer for attention to replicates and bringing attention to the relationship between input data noise and prediction accuracy. In the revised version, we highlighted our detailed analysis of replicates and explicitly compared the variation in replicates with prediction accuracy. We added the following sentences in the methods section.

“All pairwise interaction experiments (checkerboard and diagonal testing) were done in two biological replicates. The agreement between checkerboard replicates (r = 0.73, p < 0.01) or diagonal replicates (r = 0.83, p < 0.01) are given in Figure 1 or Figure 2, respectively. Our 3-drug predictions were based on diagonal scores, which were significantly but not perfectly correlated. To test predictions for 3-drugs, we calculated the correlation between predictions and empirical interaction scores. Importantly, the correlation between predictions and experiments cannot be higher than the correlation among experiments. Therefore, the highest correlation we may expect from prediction-experiment comparison is 0.83 (correlation among replicates of pairwise diagonal experiments). The correlation of 0.5 between prediction-experiment was able to capture 0.5/0.83*100 = 60% of the predictable correlation despite the underlying noise in the data.”

3. The authors predicted higher order drug-drug interactions based on the average of paired drug interactions. They explained their strategy as “For all three-drug combinations of these 8 compounds, we generated predictions by using the arithmetic mean of interaction scores for all 3 pairwise combinations among 3 drugs. For combination A+B+C, the mean (AB, AC, BC) provides the expectation for A+B+C if there is no additional synergy/antagonism associated with the three-drug combination of these drugs”. This is an interesting yet extremely simple method. This approach needs better explanation/justification. How did the authors chose this simple approach, what are the examples in the literature, how does their method compare to others? Even If they do not provide a detailed benchmark, a good discussion and scientific justification would strengthen the paper.

We thank the reviewer for the opportunity to clarify this point. It has been previously published that the mean of pairwise scores provide reliable estimates for a 3-drug interaction score. This is explained by the factorization of a high-order interactions. In the introduction of the revised submission, we included one additional paragraph to describe the rationale behind our prediction method. We referred to this paragraph in the results section when the method is applied.

Additional paragraph in introduction:

“When three drugs A, B, and C are combined, the observed (nominal) interaction has four components: Three pairwise interactions (A+B, A+C, B+C) and the interaction resulting specifically from the combination of all three components (A+B+C). This specific interaction resulting only from the high-order combination has been referred to as “emergent interaction.” These four components can be expressed as: nominal interaction = emergent interaction + mean(three pairwise interactions). Moreover, multiple studies have shown that emergent interactions are rare. Therefore, simple mean of pairwise interactions may provide a reliable estimate for nominal three-drug interaction scores.”

Edited results:

“For all three-drug combinations of these 8 compounds, we generated predictions by using the arithmetic mean of interaction scores for all 3 pairwise combinations among 3 drugs, as introduced above. For combination A+B+C, the mean (AB, AC, BC) provides the expectation for A+B+C if there is no additional synergy/antagonism associated with the three-drug combination of these drugs”.

4. The prediction power seems to be sufficiently strong based on the analysis in the manuscript. Yet we do not know about the specificity f the predictions. The authors should take a bootstrapping approach to test the predictive power of shuffled data. The predictions from shuffled data will generate a null model of predictions and statistical comparison of their actual predictions to this null model can provide better insight on the value of the predictions.

We thank the reviewer for this suggestion. In the revised submission, we include an analysis using shuffled data to test specificity, adding the following sentences to the results section:

“To test specificity, we used shuffled pairwise interaction data to predict 3-drug interaction scores. In 100,000 simulations, predictions derived from shuffled pairwise data sets showed a correlation equal to or more than real data in 3% of the cases. In addition, we used real pairwise interaction data to predict shuffled 3-drug interaction scores. In 100,000 simulations, predictions showed a correlation equal or more than real data in 1.2% of the cases. The corresponding p-values 0.03 and 0.01 attest to the specificity of our predictions.”

Attachment

Submitted filename: ResponseToReviewers_061520.docx

Decision Letter 1

Luis Eduardo M Quintas

25 Jun 2020

Guided screen for synergistic three-drug combinations

PONE-D-20-08982R1

Dear Dr. Cokol,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice for payment will follow shortly after the formal acceptance. To ensure an efficient process, please log into Editorial Manager at http://www.editorialmanager.com/pone/, click the 'Update My Information' link at the top of the page, and double check that your user information is up-to-date. If you have any billing related questions, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Luis Eduardo M Quintas, Ph.D.

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #1: All comments have been addressed

Reviewer #3: All comments have been addressed

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #3: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #3: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #3: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #3: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: Response to the reviewers' comments acceptable and I think the manuscript is now suitable for publication.

Reviewer #3: Thank you for addressing all the points. I think the article is now ready for publication.

The article is now statistically more rigorous.

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #3: No

Acceptance letter

Luis Eduardo M Quintas

30 Jun 2020

PONE-D-20-08982R1

Guided screen for synergistic three-drug combinations

Dear Dr. Cokol:

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now with our production department.

If your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information please contact onepress@plos.org.

If we can help with anything else, please email us at plosone@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Luis Eduardo M Quintas

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    Attachment

    Submitted filename: ResponseToReviewers_061520.docx

    Data Availability Statement

    Experimental data obtained during this study and calculated interaction scores are available from: https://doi.org/10.6084/m9.figshare.12043626.


    Articles from PLoS ONE are provided here courtesy of PLOS

    RESOURCES