Next Article in Journal / Special Issue
Acute and Chronic Toxicity of Carbamazepine on the Release of Chitobiase, Molting, and Reproduction in Daphnia similis
Previous Article in Journal
Museum Moving to Inpatients: Le Louvre à l’Hôpital
Previous Article in Special Issue
Methods for Evaluating the Combined Effects of Chemical and Nonchemical Exposures for Cumulative Environmental Health Risk Assessment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Investigation of the Synergistic Toxicity of Binary Mixtures of Pesticides and Pharmaceuticals on Aliivibrio fischeri in Major River Basins in South Korea

1
Environmental Safety Group, Korea Institute of Science and Technology (KIST) Europe, Campus E 7.1, 66123 Saarbruecken, Germany
2
Center for Bioinformatics, Saarland University, Campus E 2.1, 66123 Saarbruecken, Germany
3
Division of Energy and Environment Technology, KIST School, University of Science and Technology, Hwarang-ro 14-gil, Seoul 02792, Korea
4
Chemical Safety Research Center, Korea Research Institute of Chemical Technology (KRICT), 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Korea
*
Author to whom correspondence should be addressed.
Int. J. Environ. Res. Public Health 2019, 16(2), 208; https://doi.org/10.3390/ijerph16020208
Submission received: 6 December 2018 / Revised: 2 January 2019 / Accepted: 10 January 2019 / Published: 13 January 2019

Abstract

:
This work introduces the potential synergistic toxicity of binary mixtures of pesticides and pharmaceuticals, which have been detected in substantial amounts in major river basins in South Korea. Different dose-response curve functions were employed in each experimental toxicity dataset for Aliivibrio fischeri. We tested the toxicity of 30 binary mixtures at two effect concentrations: high effect concentration [EC50] and low effect concentration (EC10) ranges. Thus, the toxicological interactions were evaluated at 60 effected concentration data points in total and based on model deviation ratios (MDRs) between predicted and observed toxicity values (e.g., three types of combined effects: synergistic (MDR > 2), additive (0.5 ≤ MDR ≤ 2), and antagonistic (MDR < 0.5)). From the 60 data points, MDRs could not be applied to 17 points, since their toxicities could not be measured. The result showed 48%-additive (n = 20), 40%-antagonistic (n = 17), and 12%-synergistic (n = 6) toxicity effects from 43 binaries (excluding the 17 combinations without MDRs). In this study, EC10 ratio mixtures at a low overall effect range showed a general tendency to have more synergistic effects than the EC50 ratio mixtures at a high effect range. We also found an inversion phenomenon, which detected three binaries of the combination of synergism at low concentrations and additive antagonism at high concentrations.

1. Introduction

Conventional chemical risk assessments frequently focus on individual substances rather than mixtures even though previous studies have shown that different mixture toxicities can results from combined effects among chemicals, even at no observed effect concentrations [1,2]. The types of the mixture toxicity are generally explained as additivity, synergism (greater than additivity), and antagonism (less than additivity) [3,4]. Among these combined effects, additive and synergistic toxicity effects can be regarded as more significant than the antagonistic effect (from the aspect of the regulatory chemical risk assessment, which assumes the worst-case scenario as the default) [5]. In this context, the conventional regulatory risk assessment of chemical mixtures is mostly based on the concentration addition (CA) model as a default for estimating the mixture toxicity [6,7,8,9]. Although the worst-case scenario can be substantial synergistic toxicity, the CA model, which ignores the synergistic interaction, has been mainly employed since available predictive models for estimating the synergistic effect have been very limited for the purpose of regulatory risk assessment [5]. In addition, some studies have shown that the synergistic interaction could be a relatively rare occurrence, at least within pesticide mixtures and realistic mixtures having low concentrations in mammals (which are comprised of approximately 5% of the tested mixtures) [10,11,12,13]. In contrast, a recently published review showed that less than 25% of research and experiments on the toxicity of chemical mixtures investigated seven or more compounds [14,15]. Some clinical researchers in human toxicology also showed that the probability of synergistic toxicity could be increased in proportion to the number of components, e.g., an 8% toxicity effect was caused by pharmaceutical products with five to ten mixed components, and a 38% toxicity effect was provoked by pharmaceuticals with fourteen or more mixture components [5,16,17]. This issue related to synergism is still controversial and difficult to conclude since most of the studies concerning mixture toxicity have been conducted with a specific binary mixture or simple mixtures having less than ten components [5]. Many studies have found that pesticides and pharmaceuticals were detected in the aquatic environment and thus they might lead to mixture toxicity for aquatic nontarget organisms [14,18,19,20,21]. Thus, when considering environmental mixtures with complex matrices and different chemicals, any potential risk posed might have been underestimated by the CA model if the synergistic interaction occurred in such mixtures [14].
The objective of this study was to preliminarily investigate the potential synergistic interactions of pharmaceutical and pesticide residues that can be found in the aquatic environment. For this purpose, we tested and evaluated the toxicity of different binary mixtures of pesticides and pharmaceuticals, which had been substantially identified in major river basins in South Korea. To evaluate the toxicity of single and mixed chemicals, a bioassay with a luminescent bacterium, Aliivibrio fischeri, which is widely used in standard toxicity methods, was conducted [22,23]. Based on the original best-fit approach [24], different dose-response curve (DRC) functions were employed to test data sets, and best-fit functions of single and mixed chemicals were used in the mixture toxicity modeling.

2. Materials and Methods

2.1. Selection of Target Pesticides and Pharmaceuticals

Through previously published studies [25,26,27,28,29,30,31] that investigated the environmental concentration level of 47 pharmaceuticals in four major river basins (Han River, Geum River, Bukhan River, and Yeongsan River) in South Korea, 29 pharmaceuticals could be identified [32]. In addition, based on a study by Lee et al. [33] monitoring 140 pesticide residues in six Korean river basins (Han River, Geum River, Bukhan River, Yeongsan River, Mankyeong River, and Seomjin River), eight pesticides (isoprothiolane > butachlor > prothiofos > chlorpyrifos > hexaconazole molinate > diazinon and alachlor) could be found. From those 29 pharmaceuticals and eight pesticides, six chemicals, including four pharmaceuticals (tetracycline, trimethoprim, sulfamethoxazole, and chlortetracycline) and two pesticides (hexaconazole, and isoprothionlane), were finally selected as target mixutre components in this study (Table S1). The selction was made by considering their toxicities to A. fischeri, solubilities in water and carrier solvents, and detection frequencies in the aquatic environment. To our knowledge, this is the first study to investigate the toxicity of hexaconazole, isoprothiolane, and chlortetracycline and their binary mixtures to A. fischeri.

2.2. Test Reagents, Chemicals and Sample Preparation

Six target compounds were purchased from Sigma-Aldrich (Seelze, Germany). According to the physico-chemical properties of these compounds, stock solutions were prepared in either 99.9% dimethyl sulfoxide (DMSO, Sigma-Aldrich, Seelze, Germany) for trimethoprim and chlortetracycline or 99.8% ethanol (EtOH, Carl Roth GmbH, Karlsruhe, Germany) for tetracycline, hexaconazole, isoprothiolane and sulfamethoxazole. All of the stock solutions were kept at −20 °C under dark conditions until the working solutions were prepared. For quality control and quality assurance, the concentrations of the stock solutions were quantified with an HPLC system (Agilent 1290, Agilent Technologies, Santa Clara, CA, USA) connected to an Agilent triple quadrupole mass spectrometry (MS/MS) model 6460. Before the experiments, working solutions (1:25) were prepared by diluting the stock solutions in 2% sodium chloride for a marine bacterium, A. vibrio, according to ISO 11348-3 [22]. The pH values of the working solutions were checked and adjusted to between 6.0 and 8.0 with 1 N NaOH and 1 N HCl.

2.3. Testing Organism and Culture

The bioluminescent bacteria A. fischeri (strain NRRL-B-11177 and formerly called Vibrio fischeri) were purchased from MicroTox® (Lot number 15C4025A, Modern Water, New Castle, UK). The freeze-dried bacteria were activated with the reconstitution solution provided by the MicroTox® for 30 mins at 15 °C. The activated bacteria were transferred to a photobacteria medium (Sigma-Aldrich) for preculture at 20 °C. For stock culturing, activated A. fischeri were estimated in a 250 ml main culture medium at an initial turbidity of a 1:10 dilution by UV-vis photometric (Ultraspec 3300, Amersham, Buckinghamshire, UK) at OD578 was 0.02 (10 formazine turbidity units, FNU). A. fischeri were cultured at 20 °C with shaking at 180 rpm/min until the turbidity of the OD578 was 1.74 (700 FNU to 1800 FNU). The amplified A. fischeri were purified twice with a 2% sodium chloride solution at 4 °C and 20 min at 7000 × g. The bacteria were slowly suspended in protective medium (66 g D-(+)-glucose monohydrate, 4 g sodium chloride, 2 g L-histidine and 0.5 g BSA in 100 mL) at an ice cooled condition until the turbidity of the OD578 was 2.58 (2000 FNU to 3000 FNU). The suspended stock bacteria were stored at −80 °C.

2.4. Single Chemical Toxicity Test

Determination of an effective concentration of samples was performed [34] using the standardized methods of ISO 11348-3, 1998 by luminescent bacteria (A. fischeri) [22]. To activate, the frozen bacteria were suspended using a reconstituted solution (20 g sodium chloride, 0.3 g potassium chloride, HEPES 50 mM and glucose 50 mM for 1 liter) for 30 minutes at 15 °C. The activated luminescence bacteria were mixed with 2% sodium chloride at a 1:25 dilution. Six single compounds were serially diluted at a ratio of 1:1 (100 µL) on a flat-bottomed black 96-well plate (Greiner Bio-one, Kremsmünster, Germany). The bacterial suspensions were exposed to the serially diluted sample at a ratio of 100 µL by 100 µL. Then, reactive samples were measured after 15 min of exposure at 15 °C by a luminescent reader (Tristar2, Berthold technologies, Bad Wildbad, Germany). To assure the quality of the bacteria, a 100 ppm zinc sulfate solution (Sigma-Aldrich) was measured every time.

2.5. Mixture toxicity test

An investigation of the synergistic toxicity of all binary mixtures that could be prepared from the six target compounds in Table S1 was conducted in a fixed ratio design based on the equitoxic mixture and the generation of dose-response curves from the mixtures employed in previous studies [35,36,37,38,39]. The advantages of the fixed ratio design are not only the ability to maximize the distribution of the effective dose range but also to minimize the number of experiments [37,40,41,42]. With the same compounds in Table S1, two different equitoxic mixtures were prepared at a 50% effective concentration for each compound as a high effective concentration ratio mixture (EC50 ratio mixture) to A. fischeri and at a 10% effective concentration ratio mixture (EC10 ratio mixture) as a low effective concentration ratio mixture. As shown in Table 1, a total of 30 equitoxic binary mixtures of each combination were tested at high and low effective concentration levels. However, the total doses of the mixtures were systematically different.

2.6. Statistical Analysis of the Mixture Toxicity

A “best-fit” approach [24,39] was used to select the best model with the smallest sum of absolute residuals among the different sigmoidal functions because no single function could statistically describe all of the DRCs. Best-fit models with three-parameter sigmoidal equations were finally determined and applied to describe the experimental data of the mixture components tested in this study. The parameters in the sigmoidal regression equations and 95% confidence intervals were estimated using SigmaPlot® (Ver. 12.5, Systat Software, Chicago, IL, USA). The ECx (e.g., EC10 and EC50) of the test chemicals was derived from the regression models shown in Table 2 and Table S2. It was assumed that all of the models were confined to the effects range from 0 to 100%. In order to compare between the highest and lowest toxicity without, we expressed the orders of magnitude which were determined as follows:
Orders   of   magnitude = log ( E C x h i g h e s t   t o x i c i t y E C x l o w e s t   t o x i c i t y )
However, in case a test chemical showed low solubility in water, the model for that chemical was assumed to have a range between >0% and <the maximum effect (%), where the chemicals were present at a maximum solubility in water under the test conditions in this study. The ECx for mixtures was calculated by the CA model according to the Loewe equation [11,43]:
E C x mix = ( i = 1 n p i E C x i ) 1
where ECxmix is the predicted effective concentration of a mixture; Pi and ECxi are the fraction and the individual effective concentration of the component with in the mixture, respectively.
MDR values (Belden et al., 2007 [11]) were used to quantify the interaction between the mixture components. The MDR values were frequently applied to determine the type of interactions of the mixture toxicity [11,13,44]. MDR is defined as:
M D R = P r e d i c t e d   E C x   o f   m i x t u r e O b s e r v e d   E C x   o f   m i x t u r e
where the predicted ECx indicates the effective concentration of a mixture based on the predictive model, and the observed ECx is the effective concentration of the mixture obtained from experimental toxicity testing. In this study, the CA model, which is recommended as a default approximation for mixtures, was used to predict mixture toxicity [5,10,11,45,46]. Based on the MDR value, the types of combined effects are divided into three groups: synergistic (MDR > 2), additive (0.5 ≤ MDR ≤ 2) and antagonistic (MDR < 0.5) [11,13].

3. Results and Discussion

3.1. DRCs for Single Compounds

DRCs of all six compounds in Table S1 were empirically determined for A. fischeri, as illustrated in Figure 1.
Table 2 summarizes the parameter values of all the best-fitting regression models for the DRCs of single compounds. Table S2 explains the regression models described in Table 2. The best-fitting curves for all of the compounds showed high regression coefficients (r2) ranging from 0.969 to 0.995 and their ANOVA p-values were less than 0.0001. As presented in Table 2, the deviation between the steeper function of the highest slope values (65.25 for chlortetracycline) and the more gradual slope of the lowest (0.0034 for sulfamethoxazole) slope values was approximately 4.28 orders of magnitude. These considerable differences among the slopes of the DRCs of the compounds suggest that the compounds had highly different curve shapes for estimating the toxicity of each compound. The best-fitted DRCs for each compound had high regression coefficients (r2) ranging from 0.971 to 0.995. The EC50 values of A. fischeri were up to 0.82 orders of magnitude and ranged from 51.65 μM for hexaconazole to 338.81 μM for trimethoprim. However, the EC10 values of A. fischeri were up to 1.66 orders of magnitude and ranged from 1.05 μM for isoprothiolane to 47.79 μM for sulfamethoxazole. That is, hexaconazole and isoprothiolane presented the lowest effective concentrations (or the highest toxicity) at the respective EC50 and EC10 values, whereas isoprothiolane and sulfamethoxazole showed the highest effective concentrations (or the lowest toxicity) at the corresponding EC50 and EC10 values. These results also show that the toxicological profiles of compounds can be varied according to a given effective concentration level. In the cases of isoprothione, tetracycline, haxaconazole and trimethoprim, more than 80%-effect concentrations couldn’t be obtained under the testing conditions because of their water solubility limits.

3.2. DRCs of Binary Mixtures

The DRCs of binary mixtures in Table 1 were experimentally evaluated with high (EC50 + EC50) and low (EC10 + EC10) exposure levels (Figure 2; Figure 3). As shown in Table 3, best-fitting curves for all mixture combinations had high regression coefficients (r2) ranging from 0.817 to 0.997 except for four mixture combinations. The four exceptions are Mixture 14 (trimethoprim with hexaconazole; Mixture 14, r2 = 0.455), 18 (trimethoprim with isoprothiolane; Mixture 18, r2 = 0.429) 24 (sulfa-methoxazole with isoprothiolane; Mixture 24, r2 = 0.698) and 28 (hexaconazole with isoprothiolane; Mixture 28, r2 = 0.257).
The probable reason for the high deviation of those two mixtures is that they were the EC10 ratio mixture, i.e., an equitoxic mixture based on ratios at 10% effective concentrations for each component but significantly less toxic than the others so that the ECx values could not be appropriately determined. As tested binary mixtures, the effective concentration data ranged from a low of 2.17 µmol/L (EC10 + EC10, tetracycline and hexaconazole) to a high of 779.94 µmol/L (EC50 + EC50, trimethoprim and sulfamethoxazole).

3.3. Statistical Analysis of Mixture Toxicity to Investigate Synergism

Figure 2 and Figure 3 illustrate DRCs for the observed bioluminescent inhibitions and the predicted inhibition of A. fischeri by the CA model for the binary equitoxic mixtures based on ratios at 50% and 10% effective concentrations and following the combinations in Table 1, respectively.
To quantify the toxicity interactions between mixture components, we calculated MDR values as shown in Table 4. Based on the MDR value, we strictly divided the three types of combined effects into synergistic (MDR > 2), additive (0.5 ≤ MDR ≤ 2) and antagonistic (MDR < 0.5) [13]. As shown in Figure 2 and Table 4, ten binaries of the EC50 ratio mixtures (i.e., EC50mix) showed the same interactions at two effective concentration values of EC10 and EC50. That is, four binaries of the EC50mix (Mixture 1, 5, 9, and 25) showed the additive effects at the EC50 and EC10 ranges. The antagonistic interaction of six binaries of EC50mix (Mixture 13, 17, 19, 23, 27, and 29) was then detected at the EC50 and EC10 ranges. Interestingly, five binaries of the mixture combination detected different interactions from the EC50 and EC10 ranges. Two binaries of the EC50mix (Mixture 3 and 15) found an additive interaction at the EC50 ranges and it resulted in a synergistic interaction at the EC10 ranges. At similar trend in one binary combination (Mixture 11) was observed for the antagonistic interaction at the EC50 ranges and an additive interaction at the EC10 ranges. In contrast, the two binaries of the EC50mix (Mixture 7, and 21) resulted in opposite trends in additive interaction at the EC50 ranges and antagonistic interaction at the EC10 ranges. These observed inversion phenomena of interaction between synergism at low concentrations and additive or antagonism at high concentrations are difficult to explain. As shown in Table 4, similar phenomena have been reported in previous studies [47,48,49,50]. Wang et al. [47] tested the spiramycin and ampicillin antibiotics on Microcystis aeruginosa at different equitoxic ratios. The study found the equivalent ratio (1:1) of the binary mixture of spiramycin and ampicillin showed a synergistic interaction at low concentrations and an antagonistic interaction at high concentrations.
In a study Nica et al. [48], they tested five veterinary pharmaceuticals for the interaction of synergistic, additive and antagonistic effects on A. fischeri by the combination index of the CA and IA models. They found inversion phenomena of antagonism at high concentrations and synergism at low concentrations from six binary combinations and one pentanary mixture with individual predicted no-effect concentrations. The authors assumed that these phenomena seemed to be independent of the mode of action, which are likely complex and mostly unknown in nature. Rodea-Palomares et al. [49] also found interesting results, i.e., opposite interactions between different aquatic organisms of cyanobacteria (Anabaena CPB 4337) and A. fischeri for three pharmaceuticals. The authors reported a tandemly changing interaction from antagonistic and additive effects at low effective concentration levels to synergistic effects at high effective concentration levels in an A. fischeri test for binary and tertiary mixtures.
As shown in Table 5, Anabaena tests showed a converse pattern against A. fischeri toxicity results. Ismael et al. carefully assumed that pharmaceuticals were shared by a common binding motif such as the same target and receptor sites. Because of the structural similarity, these unexpected interactions were shown between different aquatic organisms. Gonzalez-Pleiter et al. [50] also tested cyanobacteria (Anabaena CPB 4337) for levofloxacin and tetracycline. They found synergism at low effective concentration levels and antagonism at high effective concentration levels. Based on these results, we assume that the unexpected interaction was caused by tetracycline. In some cases, the interactions of experimental toxicity screening were different from predictive toxicity results with inversion phenomena of the interaction [47,48,49,50]. This observation suggests that the binary components could make the synergistic effects at low concentrations. Wang et al. [51] reported that the reaction mechanism differs between short-term (acute) and long-term (chronic) exposures because of quorum sensing molecules which known as ain and lux in A. fischeri [51]. So that better understanding the accurate synergistic interaction, chronic toxicity tests are required. In this study, the exposure levels of the tested chemicals were less than their environmentally relevant concentrations. Thus, further studies are needed to determine how substances as synergists interact with biomolecules at low concentrations from different model organisms including A. fischeri.
As shown in Figure 3 and Table 4, the reason for not available (i.e., n.a.) data indicates that they did not reach the EC50 and EC10 ranges. Thus, all binaries of the EC10 ratio mixtures (i.e., EC10mix) were not calculated for interactions from the binary mixture at EC50 ranges. Two binaries of EC10mix (Mixture 14 and 18) did not reach the experimental data at the EC10 ranges. Four binaries of the EC10mix (Mixture 2, 4, 16, and 30) showed synergistic interactions at the EC10 ranges, whereas seven additive interactions of EC10mix (Mixture 6, 8, 10, 12, 20, 22, and 26) and two antagonistic interactions of EC10mix (Mixture 24, and 28) occurred at EC10 ranges.
Figure 4 illustrates the cumulative distribution of MDRs. In total, the binary combinations of pharmaceuticals and pesticides detected in major river basins in Korea showed 48%-additive (n = 20 from Table 4 excluding combinations without MDR), 40%-antagonistic (n = 17), and 12%-synergistic (n = 6) toxicity effects from 43 binaries on the basis of the MDR values at high (EC50) and low effect (EC10) ranges. In this study, the EC10 ratio mixtures were at a low overall effect range and showed a general tendency to have more synergistic effects than EC50 ratio mixtures at the high effect range.

4. Conclusions

In this study, the toxicity of the six target compounds (e.g., four pharmaceuticals and two pesticides) detected in major river basins in South Korea and their binary mixtures (30 samples) were tested at high and low effect concentrations (e.g., EC50 and EC10 ratio mixtures) with luminescent bacterium A. fischeri. Thus, their toxicological interactions were evaluated at 60 effect concentration data points in total and based on model deviation ratios (MDRs) between predicted and observed toxicity values. The mixture toxicities of these mixtures were also predicted by the CA model to evaluate their toxicological interactions (e.g., additive, synergistic, and antagonistic effects) based on the MDR value. From the 60 data points, MDRs were not possible for 17 points since their toxicities could not be measured. The result showed 48%-additive (n = 20), 40%-antagonistic (n = 17), and 12%-synergistic (n = 6) toxicity effects from 43 binaries (excluding 17 combinations without MDRs). That is, from the mixture toxicity evaluation and modeling, we found twenty combinations of additive effects, seventeen combinations of antagonistic effects and six combinations of synergistic effects. In addition, we found inversion phenomena such as synergism at low concentrations and additive antagonism at high concentrations. The exposure levels of the tested chemicals were less than their environmentally relevant concentrations. Since the environmentally relevant concentrations of pesticides and pharmaceuticals detected in the aquatic environment can be present at low concentrations, further studies with different species need to be conducted to clarify the mechanisms, which can address what creates these inversion phenomena.

Supplementary Materials

The following are available online at https://www.mdpi.com/1660-4601/16/2/208/s1, Table S1: Selected pesticides and pharmaceuticals, which were identified in major river basins in South Korea, Table S2: The regression models employed in describing the dose-response curves in this study.

Author Contributions

Y.K., J.K. and I.-H.B. conceived and designed the experiments. I.-H.B. performed the experiments. J.K. analyzed the best-fit curves and modeled the mixture toxicity. S.B. mass analyzed the stock solution. I.-H.B. contributed the reagents/materials/analysis tools. I.-H.B. and J.K. wrote the paper, and Y.K. proofread the manuscript.

Funding

This study was supported by the National Research Council of Science & Technology(NST) grant by the Korea government (MSIP) (No. CAP-17-01-KIST Europe) and Korea Institute of Science and Technology Europe (KIST-Europe) joint program (Project no. 11805).

Acknowledgments

We would like to thank Sungdo Lee for help with the experimental setup.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kortenkamp, A.; Altenburger, R. Approaches to assessing combination effects of oestrogenic environmental pollutants. Sci. Total Environ. 1999, 233, 131–140. [Google Scholar] [CrossRef]
  2. Junghans, M.; Backhaus, T.; Faust, M.; Scholze, M.; Grimme, L.H. Application and validation of approaches for the predictive hazard assessment of realistic pesticide mixtures. Aquat. Toxicol. 2006, 76, 93–110. [Google Scholar] [CrossRef] [PubMed]
  3. Plackett, R.L.; Hewlett, P.S. Quantal responses to mixtures of poisons. J. R. Stat. Soc. Ser. B (Methodol.) 1952, 14, 141–163. [Google Scholar] [CrossRef]
  4. German Federal Environment Agency. Ecotoxicological Combined Effects from Chemical Mixtures Part 1—Relevance and Adequate Consideration in Environmental Risk Assessment of Plant Protection Products and Biocides; Umweltbundesamt: Dessau-Roßlau, Germany, 2014. [Google Scholar]
  5. Kim, J.; Kim, S.; Schaumann, G.E. Reliable predictive computational toxicology methods for mixture toxicity: Toward the development of innovative integrated models for environmental risk assessment. Rev. Environ. Sci. Bio-Technol. 2013, 12, 235–256. [Google Scholar] [CrossRef]
  6. European Commission. State of the Art Report on Mixture Toxicity-Final Report; The School of Pharmacy, University of London: London, UK, 2009; pp. 1–31. [Google Scholar]
  7. US EPA. Guidelines for the Health Risk Assessment of Chemical Mixtures; US Environment Protection Agency: Washington, DC, USA, 1986; pp. 1–29.
  8. US EPA. Technical Support Document on Health Risk Assessment of Chemical Mixtures. Office of Research and Developmen; US Environment Protection Agency: Washington, DC, USA, 1990.
  9. US EPA. Supplementary Guidance for Conducting Health Risk Assessment of Chemical Mixtures. Risk Assessment Forum Technical Pannel; US Environment Protection Agency: Washington, DC, USA, 2000.
  10. Deneer, J.W. Toxicity of mixtures of pesticides in aquatic systems. Pest Manag. Sci. 2000, 56, 516–520. [Google Scholar] [CrossRef]
  11. Belden, J.B.; Gilliom, R.J.; Lydy, M.J. How well can we predict the toxicity of pesticide mixtures to aquatic life? Integr. Environ. Assess. Manag. 2007, 3, 364–372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Cedergreen, N.; Cristensen, A.M.; Kamper, A.; Kudsk, P.; Mathiassen, S.K.; Streibig, J.C.; Sørensen, H. A rivew of independent action compared to concentration addition as reference models for mixtures of compounds with different molecular target sites. Environ. Toxicol. Chem. 2008, 27, 1621–1632. [Google Scholar] [CrossRef] [PubMed]
  13. Cedergreen, N. Quantifying synergy: A systematic review of mixture toxicity studies within environmental toxicology. PLoS ONE 2014, 9, e96580. [Google Scholar] [CrossRef]
  14. Heys, K.A.; Shore, R.F.; Pereira, M.G.; Jones, K.C.; Martin, F.L. Risk assessment of environmental mixture effects. RSC Adv. 2016, 6, 47844–47857. [Google Scholar] [CrossRef] [Green Version]
  15. Kortenkamp, A.; Evans, R.; Faust, M.; Kalberlah, F.; Scholze, M.; Schuhmacher-Wolz, U. Investigation of the state of the science on combined actions of chemicals in food through dissimilar modes of action and proposal for science-based approach for performing related cumulative risk assessment. EFSA Support. Publ. 2012, 9. [Google Scholar] [CrossRef] [Green Version]
  16. Smith, J.W.; Seidle, L.G.; Cluff, L.E. Studies on the epideminology of adverse drug reaction (v); clinical factors influencing susceptibility. Ann. Intern. Med. 1966, 65, 629–640. [Google Scholar] [CrossRef] [PubMed]
  17. Cho, H.Y.; Lee, Y.B. Mechanism-Based Drug Interactions; Shinilbooks: Seoul, Korea, 2005. [Google Scholar]
  18. Boxall, A.B.A. The environmental side effects of medication. EMBO Rep. 2004, 5, 1110–1116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Ebele, A.J.; Abou-Elwafa Abdallah, M.; Harrad, S. Pharmaceuticals and personal care products (PPCPs) in the freshwater aquatic environment. Emerg. Contam. 2017, 3, 1–16. [Google Scholar] [CrossRef]
  20. Jeong, Y.; Schaffer, A.; Smith, K. A comparison of equilibrium and kinetic passive sampling for the monitoring of aquatic organic contaminants in German rivers. Water Res. 2018, 145, 248–258. [Google Scholar] [CrossRef] [PubMed]
  21. Battaglin, W.A.; Bradley, P.M.; Iwanowicz, L.; Journey, C.A.; Walsh, H.L.; Blazer, V.S. Pharmaceuticals, hormones, pesticides, and other bioactive contaminants in water, sediment, and tissue from rocky mountain national park, 2012–2013. Sci. Total Environ. 2018, 643, 651–673. [Google Scholar] [CrossRef] [PubMed]
  22. ISO 11348-3. Water Qualityddetermination of the Inhibitory Effect of Water Samples on the Light Emission of Vibrio Fischeri (Luminescent Bacteria Test)—Part 3: Method Using Freeze-Dried Bacteria; European Committee for Standardization: Brussels, Belgium, 1998. [Google Scholar]
  23. ASTM D5660-96. Standard Test Method for Assessing the Microbial Detoxification of Chemically Contaminated Water and Soil Using a Toxicity Test with a Luminescent Marine Bacterium; American Society for Testing and Materials: West Conshohocken, PA, USA, 2009. [Google Scholar]
  24. Scholze, M.; Boedeker, W.; Faust, M.; Backhaus, T.; Altenburger, R.; Grimme, L.H. A general best-fit method for concentration-response curves and the estimation of low-effect concentrations. Environ. Toxicol. Chem. 2001, 20, 448–457. [Google Scholar] [CrossRef] [PubMed]
  25. Kim, M.H.; Park, J.I.; Kim, Y.H.; Choi, K.H. Prioritizing human use antibiotics for environmental health management and estimating their environmental concentrations in Korean waterway. Korean Soc. Environ. Health 2006, 10, 462–468. [Google Scholar]
  26. Korean National Institute of Environmental Research. Development of Analytical Method and Study of Exposure of Pharmaceuticals and Personal Care Products in Environment; Korean National Institute of Environmental Research: Seoul, Korea, 2006. [Google Scholar]
  27. Korea Food and Drug Administration. Human Exposure Assessment for Antibiotics in the Water Environment; Korea Food and Drug Administration: Seoul, Korea, 2007. [Google Scholar]
  28. Korean National Institute of Environmental Research. Development of Analytical Method and Study of Exposure of Pharmaceuticals and Personal Care Products in Environment (II); Korean National Institute of Environmental Research: Seoul, Korea, 2007. [Google Scholar]
  29. Park, J.; Kim, M.-H.; Choi, K.; Kim, Y.; Kim, M.-Y. Environmental Risk Assessment of Pharmaceutical: Model. Application for Estimating Pharmaceutical Exposures in the Han River Basin; Korea Environment Institute: Seoul, Korea, 2007. [Google Scholar]
  30. Choi, K.; Kim, Y.; Jung, J.; Kim, M.H.; Kim, C.S.; Kim, N.H.; Park, J. Seasonal variations of several pharmaceutical residues in surface water and sewage treatment plants of Han river, Korea. Sci. Total Environ. 2008, 405, 120–128. [Google Scholar] [CrossRef]
  31. Kim, J.-H.; Park, C.-K.; Kim, M.-Y.; Ahn, S.-G. Contamination of verterinary antibiotics and antimirobials in Han river basin. J. Korean Soc. Environ. Anal. 2008, 11, 109–118. [Google Scholar]
  32. Korean National Institute of Environmental Research. Environmental Risk Assessment of Pharmaceutical Residues; Korean National Institute of Environmental Research: Seoul, Korea, 2008. [Google Scholar]
  33. Lee, J.-H.; Park, B.-J.; Kim, J.-K.; Kim, W.-I.; Hong, S.-M.; Im, G.-J.; Hong, M.-K. Risk assessment for aquatic organisms of pesticides detected in water phase of six major rivers in Korea. Korean J. Pestic. Sci. 2011, 15, 48–54. [Google Scholar]
  34. Könemann, H. Fish toxicity tests with mixtures of more than two chemicals: A proposal for a quantitative approach and experimental results. Toxicology 1981, 19, 229–238. [Google Scholar] [CrossRef]
  35. Altenburger, R.; Backhaus, T.; Boedeker, W.; Faust, M.; Scholze, M.; Grimme, L.H. Predictability of the toxicity of multiple chemical mixtures to vibrio fischeri: Mixtures composed of similarly acting chemicals. Environ. Toxicol. Chem. 2000, 19, 2341–2347. [Google Scholar] [CrossRef]
  36. Backhaus, T.; Altenburger, R.; Boedeker, W.; Faust, M.; Scholze, M.; Grimme, L.H. Predictability of the toxicity of a multiple mixture of dissimilarly acting chemicals to vibrio fischeri. Environ. Toxicol. Chem. 2000, 19, 2348–2356. [Google Scholar] [CrossRef]
  37. Payne, J.; Rajapakse, N.; Wilkins, M.; Kortenkamp, A. Prediction and assessment of the effects of mixtures of four xenoestrogens. Environ. Health Perspect. 2000, 108, 983–987. [Google Scholar] [CrossRef] [PubMed]
  38. Wang, Z.; Chen, J.; Huang, L.; Wang, Y.; Cai, X.; Qiao, X.; Dong, Y. Integrated fuzzy concentration addition-independent action (IFCA-IA) model outperforms two-stage prediction (TSP) for predicting mixture toxicity. Chemosphere 2009, 74, 735–740. [Google Scholar] [CrossRef] [PubMed]
  39. Kim, J.; Kim, S.; Schaumann, G.E. Development of a partial least squares–based integrated addition model for predicting mixture toxicity. Hum. Ecol. Risk Assess. 2014, 20, 174–200. [Google Scholar] [CrossRef]
  40. Fang, C.J.; Duan, C.Y.; He, C.; Han, G.; Meng, Q.J. A supramolecular analog of cyclohexane sustained by aromatic c-h center dot center dot center dot pi interactions between ferrocene moieties: Molecular packing of ferrocene-containing thiosemicarbazato metal complexes. New J. Chem. 2000, 24, 697–701. [Google Scholar]
  41. Liang, Y.Z.; Fang, K.T.; Xu, Q.S. Uniform design and its applications in chemistry and chemical engineering. Chemom. Intell. Lab. Syst. 2001, 58, 43–57. [Google Scholar] [CrossRef]
  42. Zhang, Y.H.; Liu, S.S.; Liu, H.L.; Liu, Z.Z. Evaluation of the combined toxicity of 15 pesticides by uniform design. Pest Manag. Sci. 2010, 66, 879–887. [Google Scholar] [CrossRef]
  43. Loewe, S.; Muischnek, H. Über Kombinationswirkungen i. Mitteilung: Hilfsmittel der Fragestellung. Naunyn-Schmiedebergs Arch. Exp. Pathol. Pharmakol. 1926, 114, 313–326. [Google Scholar] [CrossRef]
  44. Wieczerzak, M.; Kudłak, B.; Yotova, G.; Nedyalkova, M.; Tsakovski, S.; Simeonov, V.; Namieśnik, J. Modeling of pharmaceuticals mixtures toxicity with deviation ratio and best-fit functions models. Sci. Total Environ. 2016, 571, 259–268. [Google Scholar] [CrossRef] [PubMed]
  45. Warne, M.S.J.; Hawker, D.W. The number of components in a mixture determines whether synergistic and antagonistic or additive toxicity predominate: The funnel hypothesis. Ecotoxicol. Environ. Saf. 1995, 31, 23–28. [Google Scholar] [CrossRef] [PubMed]
  46. European Commission. Opinion on Toxicity and Assessment of Chemical Mixtures; European Commission: Brussels, Belgium, 2012. [Google Scholar]
  47. Wang, Z.; Chen, Q.; Hu, L.; Wang, M. Combined effects of binary antibiotic mixture on growth, microcystin production, and extracellular release of microcystis aeruginosa: Application of response surface methodology. Environ. Sci. Pollut. Res. Int. 2018, 25, 736–748. [Google Scholar] [CrossRef] [PubMed]
  48. Di Nica, V.; Villa, S.; Finizio, A. Toxicity of individual pharmaceuticals and their mixtures to aliivibrio fischeri: Evidence of toxicological interactions in binary combinations. Environ. Toxicol Chem. 2017, 36, 815–822. [Google Scholar] [CrossRef] [PubMed]
  49. Rodea-Palomares, I.; Petre, A.L.; Boltes, K.; Leganes, F.; Perdigon-Melon, J.A.; Rosal, R.; Fernandez-Pinas, F. Application of the combination index (ci)-isobologram equation to study the toxicological interactions of lipid regulators in two aquatic bioluminescent organisms. Water Res. 2010, 44, 427–438. [Google Scholar] [CrossRef] [PubMed]
  50. Gonzalez-Pleiter, M.; Gonzalo, S.; Rodea-Palomares, I.; Leganes, F.; Rosal, R.; Boltes, K.; Marco, E.; Fernandez-Pinas, F. Toxicity of five antibiotics and their mixtures towards photosynthetic aquatic organisms: Implications for environmental risk assessment. Water Res. 2013, 47, 2050–2064. [Google Scholar] [CrossRef] [PubMed]
  51. Wang, T.; Liu, Y.; Wang, D.; Lin, Z.; An, Q.; Yin, C.; Liu, Y. The joint effects of sulfonamides and quorum sensing inhibitors on vibrio fischeri: Differences between the acute and chronic mixed toxicity mechanisms. J. Hazard. Mater. 2016, 310, 56–67. [Google Scholar] [CrossRef]
Figure 1. DRCs for the bioluminescent inhibition of A. fischeri for single compounds in Table 1 (the data points are geometric means ± standard deviation [SD] of the experimentally observed data and statistical best-fits (solid lines)).
Figure 1. DRCs for the bioluminescent inhibition of A. fischeri for single compounds in Table 1 (the data points are geometric means ± standard deviation [SD] of the experimentally observed data and statistical best-fits (solid lines)).
Ijerph 16 00208 g001
Figure 2. The DRCs for the observed bioluminescent inhibitions and the predicted inhibition (red lines) by the CA model for the binary equitoxic mixtures based on ratios at 50% effective concentrations for each component (the data points are geometric means ± standard deviation (SD) of experimentally observed data, and statistical best-fits for regression models are summarized in Table 3).
Figure 2. The DRCs for the observed bioluminescent inhibitions and the predicted inhibition (red lines) by the CA model for the binary equitoxic mixtures based on ratios at 50% effective concentrations for each component (the data points are geometric means ± standard deviation (SD) of experimentally observed data, and statistical best-fits for regression models are summarized in Table 3).
Ijerph 16 00208 g002
Figure 3. The DRCs for the observed bioluminescent inhibitions and the predicted inhibition (red lines) for the CA model for in the binary equitoxic mixtures and based on ratios at 10% effective concentrations for each component (the data points are geometric means ± standard deviation (SD)of experimentally observed data, and statistical best-fits for regression models are summarized in Table 3). The dose-response curves of 30 binary mixtures of pesticides and pharmaceuticals in Table 1, respectively.
Figure 3. The DRCs for the observed bioluminescent inhibitions and the predicted inhibition (red lines) for the CA model for in the binary equitoxic mixtures and based on ratios at 10% effective concentrations for each component (the data points are geometric means ± standard deviation (SD)of experimentally observed data, and statistical best-fits for regression models are summarized in Table 3). The dose-response curves of 30 binary mixtures of pesticides and pharmaceuticals in Table 1, respectively.
Ijerph 16 00208 g003
Figure 4. The cumulative distribution of model deviation ratios (MDRs) for quantifying the toxicity interactions of the binary mixtures of pharmaceuticals and pesticides (n = 43 from Table 4, excluding combinations without MDR) for A. fischeri (synergism: MDR > 2; additivity: 0.5 ≤ MDR ≤ 2; and antagonism: MDR < 0.5).
Figure 4. The cumulative distribution of model deviation ratios (MDRs) for quantifying the toxicity interactions of the binary mixtures of pharmaceuticals and pesticides (n = 43 from Table 4, excluding combinations without MDR) for A. fischeri (synergism: MDR > 2; additivity: 0.5 ≤ MDR ≤ 2; and antagonism: MDR < 0.5).
Ijerph 16 00208 g004
Table 1. Binary mixture designs for target pesticides and pharmaceuticals.
Table 1. Binary mixture designs for target pesticides and pharmaceuticals.
Mixture No.Substance ASubstance BMixture Design
1TetracyclineSulfamethoxazoleEC50 + EC50
2TetracyclineSulfamethoxazoleEC10 + EC10
3TetracyclineHexaconazoleEC50 + EC50
4TetracyclineHexaconazoleEC10 + EC10
5TetracyclineChlortetracyclineEC50 + EC50
6TetracyclineChlortetracyclineEC10 + EC10
7TetracyclineIsoprothiolaneEC50 + EC50
8TetracyclineIsoprothiolaneEC10 + EC10
9TetracyclineTrimethoprimEC50 + EC50
10TetracyclineTrimethoprimEC10 + EC10
11TrimethoprimSulfamethoxazoleEC50 + EC50
12TrimethoprimSulfamethoxazoleEC10 + EC10
13TrimethoprimHexaconazoleEC50 + EC50
14TrimethoprimHexaconazoleEC10 + EC10
15TrimethoprimChlortetracyclineEC50 + EC50
16TrimethoprimChlortetracyclineEC10 + EC10
17TrimethoprimIsoprothiolaneEC50 + EC50
18TrimethoprimIsoprothiolaneEC10 + EC10
19SulfamethoxazoleHexaconazoleEC50 + EC50
20SulfamethoxazoleHexaconazoleEC10 + EC10
21SulfamethoxazoleChlortetracyclineEC50 + EC50
22SulfamethoxazoleChlortetracyclineEC10 + EC10
23SulfamethoxazoleIsoprothiolaneEC50 + EC50
24SulfamethoxazoleIsoprothiolaneEC10 + EC10
25HexaconazoleChlortetracyclineEC50 + EC50
26HexaconazoleChlortetracyclineEC10 + EC10
27HexaconazoleIsoprothiolaneEC50 + EC50
28HexaconazoleIsoprothiolaneEC10 + EC10
29ChlortetracyclineIsoprothiolaneEC50 + EC50
30ChlortetracyclineIsoprothiolaneEC10 + EC10
Table 2. Parameters of the regression models for dose-response curves of A. fischeri for pesticide and pharmaceutical single compounds in Table S1 (the 95% confidence intervals are provided in the brackets).
Table 2. Parameters of the regression models for dose-response curves of A. fischeri for pesticide and pharmaceutical single compounds in Table S1 (the 95% confidence intervals are provided in the brackets).
SubstanceEC50 (μM)EC10 (μM)RM 1r2Model Parameter
A 2Β 3Γ 4
Hexaconazole51.65 (50.97–52.33)3.06 (2.38–3.74)C0.9951.43350.00350.5869
Isoprothiolane137.07 (136.14–138.0)1.05 (0.12–1.85)H0.97197.38660.33121.10 × 109
Tetracycline150.08 (148.85–151.30)10.60 (9.38–11.83)L0.9811.4806–0.7364374.5933
Trimethoprim338.81 (338.05–339.56)26.20 (25.45–26.95)L0.9901.2286–0.7997542.4834
Sulfamethoxazole254.20 (253.25–255.15)47.79 (46.83–48.74)C0.9940.95610.00341.1932
Chlortetracycline91.32 (90.52–92.12)12.32 (11.52–13.12)G0.9930.990265.254166.4673
Notes. 1 Regression models (C: Chapman, G: Gompertz, H: Hill, L: Logistic); 2 Height; 3 Slope; and 4 Center point.
Table 3. Parameters of regression models for dose-response curves of 30 binary mixtures of pesticides and pharmaceuticals in Table 1 (the 95% confidence intervals are provided in the brackets).
Table 3. Parameters of regression models for dose-response curves of 30 binary mixtures of pesticides and pharmaceuticals in Table 1 (the 95% confidence intervals are provided in the brackets).
Mixture No.EC50 (μM)EC10 (μM)RM 1r2Model Parameter
A 2Β 3Γ 4
1313.95 (313.07–314.82)16.62 (15.74–17.50)C0.9872528.44582.47 × 10−60.5477
2n.a.57.43 (6.99–7.88)G0.96819.57356.48904.8497
3133.58 (132.69–134.46)2.89 (2.01–3.78)C0.989915.21737.38 × 10−60.4200
4n.a.2.17 (1.75–2.59)G0.98323.92242.03671.8951
5129.70 (128.75–130.66)5.98 (5.02–6.93)C0.989102.00250.00250.5494
6n.a.9.54 (8.83–10.25)L0.95430.2768−0.886121.1859
7203.46 (202.88–204.04)36.83 (36.25–37.41)G0.99684.6072118.9205127.0557
8n.a.11.66 (11.25–12.06)G0.87410.41773.30481.0944
9265.16 (264.62–265.69)15.55 (15.01–16.09)C0.99699.58850.00150.6049
10n.a.8.64 (8.18–9.10)L0.97022.0641−1.43089.8530
11779.94 (779.31–780.56)62.99 (62.36–63.61)C0.98983.77650.00090.7231
12n.a.50.31 (49.80–50.81)G0.91419.204149.305429.2598
13488.97 (488.44–489.49)59.282 (58.76–59.81)H0.991131.15440.9527812.9558
14n.a.n.a.G0.4554.55230.591617.0836
15222.30 (220.95–223.65)4.56 (3.22–5.91)H0.96463.86490.763341.4132
16n.a.3.91 (3.38–4.44)C0.98833.85410.14891.4924
17637.32 (637.01–637.64)154.31 (153.99–154.63)G0.99760.7316216.8352282.2291
18n.a.n.a.G0.4298.992425.694125.7707
19209.14 (208.45–209.83)34.75 (34.06–35.44)G0.99278.4915114.7884117.7289
20n.a.26.94 (26.43–27.44)G0.81717.747230.96259.7285
21206.10 (205.57–206.64)37.41 (36.87–37.95)G0.99796.2390135.9703148.5324
22n.a.15.85 (15.31–16.38)G0.933127.428534.350616.1526
23339.80 (339.23–340.37)106.78 (106.22–107.36)G0.995183.3055163.6608229.7587
24n.a.45.77 (45.31–46.24)S0.69811804.379851.2073311.5280
25119.13 (118.31–119.96)8.40 (7.57–9.23)H0.98841.37 × 1050.60695.52 × 107
26n.a.13.89 (13.46–14.33)C0.9560158.68409.12 × 10-60.3080
27204.18 (203.53–204.83)45.75 (45.10–46.40)G0.9901107.4683139.9025166.7380
28n.a.5.82 (4.61–7.04)G0.257112.16553.7398–0.2706
29247.79 (246.98–248.59)28.44 (27.64–29.24)H0.98704.73 × 1050.74355.52 × 107
30n.a.4.38 (3.84–4.91)C0.943518.62410.11280.6594
Notes. 1 Regression models (C: Chapman, G: Gompertz, H: Hill, L: Logistic); 2 Height; 3 Slope; and 4 Center point, 5 Not available.
Table 4. Observed and predicted ECxmix values of tested mixtures of pharmaceuticals and pesticides in binary combinations, and MDR values to address the interactions between components (the 95% confidence intervals are provided in the brackets).
Table 4. Observed and predicted ECxmix values of tested mixtures of pharmaceuticals and pesticides in binary combinations, and MDR values to address the interactions between components (the 95% confidence intervals are provided in the brackets).
Mixture No.EC50mix 1EC10mix
Observed
(EC50, μM)
Predicted 2MDR 3Type 4Observed
(EC10, μM)
PredictedMDRType
EC50 ratio mixtures 5
1313.95 (313.07–314.82)169.630.54Add.616.62 (15.74–17.50)13.550.82Add.
3133.58 (132.69–134.46)128.470.96Add.2.89 (2.01–3.78)8.713.01Syn.
5129.70 (128.75–130.66)132.371.02Add.5.98 (5.02–6.93)10.921.83Add.
7203.46 (202.88–204.04)147.270.72Add.36.83 (36.25–37.41)3.840.10Anta. 7
9265.16 (264.62–265.69)189.170.71Add.15.55 (15.01–16.09)13.610.88Add.
11779.94 (779.31–780.56)281.920.36Anta.62.99 (62.36–63.61)36.510.58Add.
13488.97 (488.44–489.49)129.650.27Anta.59.282 (58.76–59.81)8.200.14Anta.
15222.30 (220.95–223.65)139.680.63Add.4.56 (3.22–5.91)16.453.61Syn. 8
17637.32 (637.01–637.64)194.160.30Anta.154.31 (153.99–154.63)2.000.01Anta.
19209.14 (208.45–209.83)101.470.49Anta.34.75 (34.06–35.44)7.210.21Anta.
21206.10 (205.57–206.64)119.870.58Add.37.41 (36.87–37.95)17.010.45Anta.
23339.80 (339.23–340.37)167.070.49Anta.106.78 (106.22–107.36)1.700.02Anta.
25119.13 (118.31–119.96)66.930.56Add.8.40 (7.57–9.23)5.040.60Add.
27204.18 (203.53–204.83)74.930.37Anta.45.75 (45.10–46.40)1.570.03Anta.
29247.79 (246.98–248.59)100.210.40Anta.28.44 (27.64–29.24)3.180.11Anta.
EC10 ratio mixtures
2n.a. 9n.a.--7.43 (6.99–7.88)17.532.36Syn
4n.a.n.a.--2.17 (1.75–2.59)8.964.13Syn.
6n.a.n.a.--9.54 (8.83–10.25)12.031.26Add.
8n.a.n.a.--11.66 (11.25–12.06)8.570.73Add.
10n.a.n.a.--8.64 (8.18–9.10)13.841.60Add.
12n.a.n.a.--50.31 (49.80–50.81)41.160.82Add.
14n.a.n.a.--n.a.n.a.--
16n.a.n.a.--3.91 (3.38–4.44)15.033.84Syn.
18n.a.n.a.--n.a.n.a.--
20n.a.n.a.--26.94 (26.43–27.44)14.000.52Add.
22n.a.n.a.--15.85 (15.31–16.38)18.521.17Add.
24n.a.n.a.--45.77 (45.31–46.24)12.980.28Anta.
26n.a.n.a.--13.89 (13.46–14.33)11.180.80Add.
28n.a.n.a.--5.82 (4.61–7.04)2.520.43Anta.
30n.a.n.a.--4.38 (3.84–4.91)10.072.30Syn.
Note. 1 ECxmix: effective concentrations of a mixture causing x% toxicity effect; 2 Values predicted by the concentration addition model; 3 Model deviation ratio; 4 Type of combined toxic effects; 5 ECx ratio mixture: an equitoxic mixture based on ratios at x% effective concentrations for each component; 6 Additivity; 7 Antagonism; 8 Synergism; and 9 Not available.
Table 5. A summary of the studies related to the interaction of inversion phenomena.
Table 5. A summary of the studies related to the interaction of inversion phenomena.
MixtureExperimental DesignSpeciesEndpointConvind EffectQuantification MethodsRef.
High LevelLow Level
Two antibioticsBinary equitoxic mixture ratio (5:1, 1:1, 1:5)Bacteria
Microcystis aeruginosa (MA)
EC50 and EC5 for MA cell from equitoxic ratio SP/Amp (5:1, 1:5, 1:1)Antagonism
1:1 ratio
>0.7 ug/L
Synergism
1:1 ratio
<0.5 ug/L
Departure from additivity model (CA, IA)[47]
Five veterinary pharmaceuticalsBinary and multicomponent mixtureBacteria
A. fischeri
Applying the combination index method from active pharmaceutical compound interactions for bacteriaAntagonismSynergismDeparture from combination index (CA, IA)[48]
Diclofenac : SulfamethizoleEC50 1.13EC10 0.61
Acetylsalycilic acid : SulfamethizoleEC50 2.58EC10 0.85
Chlortetracycline : AmoxicillinEC50 2.16EC10 0.08
Acetylsalycilic acid : DiclofenacEC50 1.13EC10 0.73
Sulfamethizole : AmoxicillinEC50 1.57EC10 0.41
Acetylsalycilic acid : AmoxicillinEC50 2.17EC10 0.72
Predicted no-effect concentration
(five pharmaceutical compound mixture)
EC50 1.36EC10 0.61
Three pharmaceuticalsBinary and ternary combinationsBacteria
A. fischeriCyanobacteria
Anabaena CPB4337
Applying the combination index with isobologram equation methods from pharmaceutical compounds for in vitro and in vivo bioassayAntagonismSynergismDeparture from combination index (CA and IA) with isobologram equation[49]
Fenofibrate : BezafibrateEC90 2.59
EC50 1.19
EC10 0.55
Fenofibrate : GemfibrozilEC90 12.9
EC50 1.29
EC10 0.13
Fenofibrate : Gemfibrozil : BezafibrateEC90 3.92EC50 0.57
EC10 0.09
Five antibioticsBinary and multicomponent mixtureCyanobacteria
Anabaena CPB4337
Microalgae
Raphidocelis subcapitata
Applying combination index with isobologram equation methods from pharmaceutical compound for in vitro and in vivo bioassayAntagonismSynergismDeparture from combination index (CA and IA) with isobologram equation[50]
Levofloxacin : TetracyclineEC50 1.6EC10 0.37

Share and Cite

MDPI and ACS Style

Baek, I.-H.; Kim, Y.; Baik, S.; Kim, J. Investigation of the Synergistic Toxicity of Binary Mixtures of Pesticides and Pharmaceuticals on Aliivibrio fischeri in Major River Basins in South Korea. Int. J. Environ. Res. Public Health 2019, 16, 208. https://doi.org/10.3390/ijerph16020208

AMA Style

Baek I-H, Kim Y, Baik S, Kim J. Investigation of the Synergistic Toxicity of Binary Mixtures of Pesticides and Pharmaceuticals on Aliivibrio fischeri in Major River Basins in South Korea. International Journal of Environmental Research and Public Health. 2019; 16(2):208. https://doi.org/10.3390/ijerph16020208

Chicago/Turabian Style

Baek, In-Hyuk, Youngjun Kim, Seungyun Baik, and Jongwoon Kim. 2019. "Investigation of the Synergistic Toxicity of Binary Mixtures of Pesticides and Pharmaceuticals on Aliivibrio fischeri in Major River Basins in South Korea" International Journal of Environmental Research and Public Health 16, no. 2: 208. https://doi.org/10.3390/ijerph16020208

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop