Next Article in Journal / Special Issue
Microbial Symbiont-Based Detoxification of Different Phytotoxins and Synthetic Toxic Chemicals in Insect Pests and Pollinators
Previous Article in Journal
Bioaccumulation of Organic and Inorganic Pollutants in Fish from Thermaikos Gulf: Preliminary Human Health Risk Assessment Assisted by a Computational Approach
Previous Article in Special Issue
Toxicity of UV Filter Benzophenone-3 in Brine Shrimp Nauplii (Artemia salina) and Zebrafish (Danio rerio) Embryos
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Review on Fluoroquinolones’ Toxicity to Freshwater Organisms and a Risk Assessment

by
Marianna Pauletto
and
Marco De Liguoro
*
Department of Comparative Biomedicine & Food Science (BCA), University of Padova, Viale dell’Università 16, I-35020 Legnaro, Padova, Italy
*
Author to whom correspondence should be addressed.
J. Xenobiot. 2024, 14(2), 717-752; https://doi.org/10.3390/jox14020042
Submission received: 21 February 2024 / Revised: 21 May 2024 / Accepted: 28 May 2024 / Published: 4 June 2024
(This article belongs to the Special Issue Environmental Toxicology and Animal Health)

Abstract

:
Fluoroquinolones (FQs) have achieved significant success in both human and veterinary medicine. However, regulatory authorities have recommended limiting their use, firstly because they can have disabling side effects; secondly, because of the need to limit the spread of antibiotic resistance. This review addresses another concerning consequence of the excessive use of FQs: the freshwater environments contamination and the impact on non-target organisms. Here, an overview of the highest concentrations found in Europe, Asia, and the USA is provided, the sensitivity of various taxa is presented through a comparison of the lowest EC50s from about a hundred acute toxicity tests, and primary mechanisms of FQ toxicity are described. A risk assessment is conducted based on the estimation of the Predicted No Effect Concentration (PNEC). This is calculated traditionally and, in a more contemporary manner, by constructing a normalized Species Sensitivity Distribution curve. The lowest individual HC5 (6.52 µg L−1) was obtained for levofloxacin, followed by ciprofloxacin (7.51 µg L−1), sarafloxacin and clinafloxacin (12.23 µg L−1), and ofloxacin (17.12 µg L−1). By comparing the calculated PNEC with detected concentrations, it is evident that the risk cannot be denied: the potential impact of FQs on freshwater ecosystems is a further reason to minimize their use.

1. Introduction

During the 1970s, a groundbreaking development emerged in the realm of pharmaceuticals with the introduction of flumequine. Scientists successfully enhanced the quinolone structure by incorporating a fluorine atom at position C6 [1]. This chemical modification not only broadened the spectrum of antimicrobial activity but also significantly improved tissue penetration. The subsequent pursuit of innovation led to the discovery of numerous other fluoroquinolones (FQs) that surpassed the efficacy of flumequine in terms of therapeutic range and pharmacokinetic properties in both humans and animals. Despite initial successes, certain FQs, including alatrofloxacin, clinafloxacin, gatifloxacin, gemifloxacin, grepafloxacin, sparfloxacin, temafloxacin, and trovafloxacin, faced swift withdrawal from the market due to severe adverse reactions [2,3]. However, other FQs have progressively emerged as the preferred choice for treating serious infections and common ailments, even though they have occasionally shown alarming side effects.
Over time, the pharmaceutical landscape evolved, and today, a diverse array of FQs exists, categorized into first, second, third, and fourth generations [4]. The development of new FQs has made an important contribution to the advancement of human medicine, with compounds such as ciprofloxacin, delafloxacin, gemifloxacin, levofloxacin, lomefloxacin, moxifloxacin, ofloxacin, pefloxacin, prulifloxacin, and rufloxacin routinely being used in healthcare settings. Additionally, veterinary medicine has benefited from dedicated FQs, such as flumequine, enrofloxacin, danofloxacin, pradofloxacin, difloxacin, marbofloxacin, sarafloxacin, and orbifloxacin [5,6].
FQs exert their antimicrobial effects by selectively inhibiting two bacterial enzymes, DNA gyrase and topoisomerase IV, which are crucial for bacterial DNA replication but absent in human cells [7]. This targeted mechanism would have made FQs specific and bactericidal, offering promising therapeutic prospects; however, research has revealed that DNA gyrases are not exclusive to bacteria, as green algae [8] and higher plants [9] also possess these enzymes. Furthermore, recent studies have shown that ciprofloxacin can interfere with type II topoisomerases (Top2α and Top2β) found in mammalian mitochondria, leading to mitochondrial damage, a phenomenon referred to as mitotoxicity [10]. Mitochondrial disruption has been associated with delayed adverse effects ranging from neurologic to musculoskeletal and cardiovascular conditions [11]. Moreover, FQs have been implicated in iron chelation, resulting in the inhibition of Fe (II)-dependent dioxygenases. This inhibition leads to DNA and histone hypermethylation, suppression of collagen prolyl hydroxylation, and inhibition of HIF mRNA translation. These molecular alterations may explain side effects, such as renal toxicity and tendinopathy [12].
As the usage of FQs in human medicine gained momentum, concerns about side effects grew. In 2015, the FDA (U.S. Food and Drug Administration) recognized FQAD (FluoroQuinolone-Associated Disability) as a syndrome, based on 178 cases where otherwise healthy individuals developed disabling and potentially irreversible conditions after taking fluoroquinolones for minor ailments. FQAD involves various symptoms that may endure even after stopping the medication. These symptoms might consist of profound tiredness, muscle frailty, joint discomfort, nerve impairment, cognitive challenges, and disturbances in mental health. Consequently, the FDA revised the boxed warning on FQs to address safety concerns [13] and advised reserving these drugs for serious infections only [2]. Similarly, the European Medicines Agency’s Pharmacovigilance Risk Assessment Committee (PRAC) recommended restricting the use of FQs and quinolone antibiotics in 2018, following reports of disabling and potentially long-lasting side effects associated with these medications [14].
In veterinary medicine, the use of FQs, particularly enrofloxacin, has also raised concerns due to reported side effects on domestic animals, affecting the skeletal and reproductive system, retinal pigment epithelium, liver enzymatic activity, and immune system [11]. These findings, combined with the risk of spreading antibacterial resistance between animals and humans, have discouraged the widespread use of FQs in both human and veterinary medicine [12]. Consequently, the once-growing trend of FQ utilization in human medicine has ended, and there is now a greater focus on judicious and restricted administration. However, although there were some modest reductions in prescriptions after the implementation of interventions in some countries, findings did not support a relevant effect of regulatory intervention on FQ use [15].
For widely used drugs like FQs, which find application in both human medicine and farm animals and are not strictly selective in their action, the potential impact on the environment has become a critical concern. FQs are known for their remarkable persistence [16], and recent scientific literature has shed light on their presence in the environment and their toxic effects, particularly on aquatic organisms [17,18]. Studies have revealed that FQs can harm both photosynthetic organisms and animals, even at low environmental concentrations. Indeed, they bear the potential to affect biodiversity through their impacts on sensitive species. Moreover, the ability of these compounds to interact with genetic material and induce delayed, reproductive, and transgenerational toxicity [19,20,21] adds to the complexity of assessing their environmental risk.
In light of these concerns, the primary objective of this paper is to provide a comprehensive and critical review of the existing knowledge regarding the presence of FQs in the aquatic environment and their toxic effects on freshwater organisms. For this purpose, a database on drug concentrations in the environment [22] was used as the main reference point for environmental contamination, whilst published data on toxic effects in freshwater organisms were retrieved based on the U.S. Environmental Protection Agency (USEPA) ecotoxicological database [23], the supplementary material of a recent review on FQs [24], and with the help of various search engines, by using the keywords “fluoroquinolones, environment, freshwater, ecotoxicity” to retrieve the relevant literature. The main criteria for selecting articles on contamination levels were the precise location of the sampling and validation of the analytical method reported; whilst for articles on toxic effects, the availability of accurate measures of acute toxicity (EC50) and/or a clear description of the mechanisms involved were discriminative. By consolidating the available information, this review aims to enhance our understanding of the potential risks posed by FQs and their impact on delicate freshwater ecosystems. Of the numerous FQs, only 11 (ciprofloxacin, clinafloxacin, enrofloxacin, flumequine, gatifloxacin, levofloxacin, lomefloxacin, moxifloxacin, norfloxacin, ofloxacin, and sarafloxacin) were considered for risk assessment, taking into account the availability of data on both their presence in the environment and toxicity to freshwater organisms. Recently, various reviews have been published on FQs in the environment [24,25,26,27,28,29]. This one may complement them by focusing on the freshwater ecosystem, giving a broad picture of the historical contamination (first quarter of the current century) in Europe, Asia, and the USA, accurately updating the mechanisms of toxicity in non-target species, and performing a novel risk assessment based on a normalized Species Sensitivity Distribution.

2. Chemistry and Mode of Action of FQs

Flumequine was the first mono-fluorinated quinolone. The addition of fluorine, in C6, to the basic quinolone structure, resulted in an increased antibacterial spectrum. This marked the beginning of intensive chemical synthesis efforts to refine structure–activity relationships and optimize pharmacokinetics. The addition of different R1, R7, and R8 groups created new and more effective FQs with broader antibacterial spectra and improved pharmacokinetics. A detailed table on the structure of the various FQs is reported in [30]. Each generation of FQs has specific advantages and disadvantages, including differences in the spectrum of activity, pharmacokinetics, side effects, and resistance patterns. According to Rusu and colleagues [31], there are four distinct generations of FQs. Of the compounds considered here for the risk assessment, flumequine belongs to the first generation; ciprofloxacin, lomefloxacin, norfloxacin, ofloxacin, and sarafloxacin to the second; clinafloxacin, enrofloxacin, and levofloxacin to the third; gatifloxacin and moxifloxacin to the fourth. FQs are used to treat a wide range of bacterial infections mainly of the urinary and respiratory tract, and are often used in cases where other antibiotics are not suitable due to resistance or allergies.
The targets of FQs are two enzymes that regulate DNA topology: gyrase and topoisomerase IV. Each of these enzymes is composed of four subunits: two GyrA and two GyrB make up the gyrase, two ParC and two ParE the topoisomerase IV. The GyrA and ParC are involved in DNA strand breakage, whilst GyrB and ParE in DNA strand assembling. Through its breaking and re-ligation processes, DNA gyrase alleviates supercoiling ahead of the replication fork, paving the way for the replication complexes to move along the DNA, whilst Topoisomerase IV, at the end of replication, removes the tangling of DNA strands, thereby allowing DNA segregation into two daughter cells. Fluoroquinolones interact with the DNA-bound enzyme to create conformational changes that result in the inhibition of normal enzyme activity. As a result, in the case of gyrase, the progression of the replication fork is blocked, thereby inhibiting normal bacterial DNA synthesis, whilst in the case of topoisomerase IV, the bacterial circular DNA is broken but no longer re-ligated [7].

3. Sources and Presence of FQs in the Environment

Wastewater from aquaculture facilities, pharmaceutical manufacturing suites, hospitals, and municipalities are major sources of watersheds contaminated with FQs. Traditional wastewater treatment plants are ineffective in removing fluoroquinolones [27]. By comparing levels in influents and effluents of 18 WWTPs around the world [32], an average removal efficiency of 64 ± 32% can be calculated for the various FQs. Additionally, a significant quota of FQs may end up in dewatered sludge [33], which is sometimes used as fertilizer, representing an additional input route into the environment. In addition, manure/slurry from large-scale animal farms where FQs are used for mass medication (prophylactic or metaphylactic) are often used as fertilizer for crop fields, contributing greatly to soil contamination [34]. Once brought onto land, FQs tend to sorb strongly at the topsoil [35]. This phenomenon prevents leaching to lower soil layers and groundwater contamination [36]; however, FQs can still reach surface water bodies through agricultural runoff [29].
In general, the adsorption of antibiotics to soils is determined by organic content, pH, cation exchange capacity, and soil texture [37]. Despite a relatively low octanol–water partition coefficient (Kow), the various FQs show a high affinity for sludge, soils, and sediments. Indeed, depending on the pH, they occur as a mixture of neutral and/or differently charged species, and electrostatic interactions may play a significant role in the sorption process [30]. FQs are known to have moderate-to-high persistence in soils and sediments, which can lead to their accumulation over time [27]. Indeed, concentrations up to a few mg kg−1 have been detected in freshwater sediments [38,39], soils fertilized with contaminated manure [34], and Waste Water Treatment Plants (WWTPs) dewatered sludge [33]. Concentrations detected in the water column have been generally in the ng to µg L−1 range [40,41], with some remarkable exceptions at the mg L−1 level, resulting from drug manufacturing activities [42,43,44] and fish farming [45] in Asian countries. In the area of the Pearl River delta (China), traces of FQs have been detected even in tap water [46,47]. In Figure 1, the highest concentrations detected in surface waters or WWTP effluents of the various countries are considered and represented by spots of different sizes according to the range they fall into.

4. Mechanisms of Toxicity of Fluoroquinolones in Non-Target Species

Once FQs have been released into the aquatic environment, they interact with the microbial communities, leading to changes in their diversity, abundance, and function. Indeed, FQs inhibit the growth of both susceptible pathogenic and beneficial bacteria, resulting in the disruption of microbial ecosystems (e.g., shift in nutrient cycling, alteration of decomposition rates) peculiar to freshwater environments [27]. In addition to this general effect, common to all antibiotics, FQs can induce a plethora of toxic effects through specific molecular mechanisms, some of which have been largely studied, whilst others have poorly so, and some have been only postulated [3]. In the present section, toxic effects and their putative molecular mechanisms of particular relevance to the freshwater species are reported. Worthy of note, molecular mechanisms have been originally investigated in mammals, and only in some cases in fish and crustaceans, but rarely studied in microalgae and aquatic plants. Thus, what we know about FQ mechanisms of toxicity mostly results from studies conducted in mammalian cells and rodents, and marginally in zebrafish and daphnids. Overall, whilst the phenotypic effects caused by FQs have been extensively studied across various freshwater species, their correlation with distinct molecular mechanisms remains inadequately explored. In this respect, mechanistic studies specifically assessing the molecular drivers of FQ toxicity should be encouraged.

4.1. Genotoxicity

The same mechanisms of action underpinning antibacterial effects of FQs might be responsible for toxicity in non-target eukaryotic organisms. Indeed, whilst initially FQs were deemed to specifically target prokaryote gyrase and topoisomerases, later on, it was discovered that enzymes with similar structures and functions might occur also in eukaryotic cells (green algae, plants, and mammals). This is the case for type II topoisomerases Top2α and Top2β, recently identified in mammalian mitochondria extracted from cultured cells and mouse tissues [10]. In particular, ciprofloxacin showed the ability to interact with the DNA-bound mammalian Top2β, thus inhibiting the enzyme activity, and resulting in the accumulation of positively supercoiled mtDNA, blockage of mitochondrial transcription and replication, reduction in mtDNA copy number, and, ultimately, impaired cell proliferation [10]. Likewise, proteomic investigations conducted in vivo in zebrafish embryos showed that ciprofloxacin and enrofloxacin decreased Top1 levels [219], most probably as a consequence of the binding between FQs and the topoisomerase, leading to enzyme degradation.
Mutagenic effects can also be caused by non-selective DNA binding of FQs that induces oxidative DNA damage (ROS overproduction), in turn causing cell death and/or accelerating cellular aging, as previously demonstrated for several drugs [220,221]. Indeed, in a recent study, the genotoxic effects of five of the FQs most used in human medicine (ciprofloxacin, levofloxacin, moxifloxacin, norfloxacin, and ofloxacin) were demonstrated through in vitro and in silico combined approaches [222]. The authors showed a discrete ability of these FQs to bind human DNA, resulting in oxidative damage and base excision repair (BER) pathway activation.
Genotoxic effects of FQs were also highlighted in vivo. In Drosophila melanogaster, ciprofloxacin and enrofloxacin were found to induce homologous recombination in proliferative cells, a DNA repair mechanism that can result in DNA rearrangements and genetic diseases [223]. In this study, homologous recombination is suggested to occur as a consequence of DNA double-strand breaks not resolved because of FQ-mediated inhibition of Top2, which affects DNA breakage/rejoining reactions. In the liver of yellow catfish Pelteobagrus fulvidraco, enrofloxacin significantly increased the level of GADD45, a protein frequently induced by DNA damage, and promoted oxidative stress and apoptosis [224]. Chronic exposure to ciprofloxacin induced genetic damage in D. magna, yet in this study oxidative stress occurred marginally, namely with a mild increase in lipid peroxidation observed [225]. Likewise, environmental concentrations of ciprofloxacin and enrofloxacin induced cellular DNA damage (i.e., DNA strand breaks) and apoptosis in zebrafish embryos, but only partly due to increased ROS [219]. Indeed, in addition to oxidative stress, other mechanisms may also contribute to DNA damage, such as the inhibition of topoisomerases, as discussed above, and the inhibition of the DNA repair machinery [219].
It is well established that the chemical structure of FQs is closely related to their genotoxic potential. Firstly, the fluorine atom in C6 improves their antibacterial efficacy but, unfortunately, also increases the genotoxicity of the compounds [226,227]. Secondly, QSAR and 3D Pharmacophore in silico models showed that the C5 and C7 positions are the main determinants of FQ genotoxicity [228]. It is worthy of note that new FQ derivatives, based on this prediction model, present lower genotoxicity and higher efficacy [228].

4.2. Degradation of the Extracellular Matrix

A further mechanism underlying FQ toxicity is the upregulation of cell matrix metalloproteinases (MMPs), sometimes coupled to the downregulation of their inhibitors (tissue inhibitor of MMPs, TIMPs). The imbalance between the production of MMPs and TIMPs leads to the degradation of collagen and elastic fibers, thus affecting the integrity of the extracellular matrix (ECM) in several tissues [229,230]. ECM disruption likely serves as a mechanism for several adverse events. For instance, in an epidemiological study conducted by the Food and Drug Administration (FDA) and the European Medicines Agency (EMA), ciprofloxacin, levofloxacin, and moxifloxacin administration were associated with potential aortic dissections or rupture [231]. Ciprofloxacin-mediated tympanic membrane healing problems might be due to a decrease in fibroblast viability and collagen and α-tubulin protein levels [232]. Likewise, ciprofloxacin and levofloxacin decreased the principal matrix protein collagen type I in human-derived tendon cells, possibly resulting in tendinopathies [233]. In addition, FQs possess chelating properties. For instance, ciprofloxacin, enrofloxacin, and norfloxacin strongly chelate the iron needed by the two enzymes crucial for collagen synthesis, prolyl 4-hydroxylase and lysyl hydroxylase, as demonstrated in human embryonic kidney cells [12]. Furthermore, it is possible that FQs may disrupt the ECM outside the retina, a tissue that contains several layers of different kinds of collagen, possibly resulting in retinal detachment. Despite this adverse effect remaining controversial, some epidemiological studies reported an association between oral administration of FQs and the risk of developing retinal detachment (e.g., [234,235]).
This mechanism of FQ toxicity which targets the ECM is poorly studied in aquatic species. Environmental concentrations of norfloxacin have recently been reported to affect zebrafish craniofacial development, putatively dysregulating the expression of genes coding for ECM proteins (e.g., collagen type II) [236]. In embryos of the same species, enrofloxacin and ciprofloxacin ≤ 0.5 μM modulate the protein level of major ECM components, namely collagens, fibronectin, and laminin [237]. Overall, based on evidence in fish and humans, we would infer that the FQ-induced dysregulation of ECM proteins might cause structural defects that may ultimately result in alterations of many tissues, with serious effects on the freshwater biota.

4.3. Toxicity to the Central Nervous System

FQs are antagonists of Gamma-Aminobutyric Acid-A (GABA-A) receptors in the brain, spinal cord, and peripheral nervous system, meaning that they displace other molecules that physiologically bind to this receptor, such as GABA. As it is an inhibitory neurotransmitter, FQs induce blockage of GABA receptors, which results in a stimulation of the central nervous system (CNS), with neurological manifestations encompassing insomnia, agitation, hallucinations, and seizures in humans. Difference in FQs’ affinity for GABA-A receptors most likely explains the variability in neurotoxic effects. Indeed, FQs containing unsubstituted 7-piperazine (e.g., ciprofloxacin, enoxacin, norfloxacin) and 7-pyrrolidine (e.g., clinafloxacin, tosufloxaxin) at their R7 position show the most significant GABA-antagonistic effects [238]. Accordingly, in zebrafish, FQs were reported to affect locomotor activity, which is a good proxy of toxicity to CNS, and a correlation between FQ structure and the exerted toxicity was also highlighted [239].
FQs may also exhibit a direct effect on excitatory brain pathways, by stimulating N-methyl-d-aspartate (NMDA) and adenosine receptors, which could lead to other CNS symptoms [240,241]. A recent study has demonstrated the occurrence of this mechanism of toxicity in zebrafish embryos exposed to FQs [242]; in the aforementioned study, developmental neurotoxicity induced by norfloxacin was demonstrated to be mediated by the activation of NMDA receptors. The involvement of these receptors in mediating FQ toxicity has been reported also in D. magna exposed to flumequine, where NMDA type I was among the top upregulated genes [19].

5. Toxicity of FQs to Freshwater Organisms

For the convenience of the reader, in Table 1, all the collected EC50s of FQs in cyanobacteria, unicellular algae, plants, crustaceans, and fish are reported and referenced, with details on duration, endpoints, and measured parameters.

5.1. Cyanobacteria

Enrofloxacin and ciprofloxacin were assayed on the cyanobacterium Anabaena flos-aquae, which was shown to be particularly sensitive, with EC50 values of 173 and 10.2 µg L−1, respectively [243]. In the same species, Gonzalez-Pleiter and co-authors reported an EC50 of 5.6 and 4.8 mg L−1 for norfloxacin and levofloxacin, respectively [244]. Robinson et al. [245] evaluated the toxicity of seven FQs on the cyanobacterium Microcystis aeruginosa; the most active compound was levofloxacin (EC50 7.9 µg L−1), whilst the least was flumequine (EC50 1.96 mg L−1). However, in the same species, Lützhøft and colleagues reported EC50s of 15 µg L−1 for sarafloxacin and 159 µg L−1 for flumequine [247]; whilst Halling-Sorensen and colleagues reported an EC50 of 5 µg L−1 for ciprofloxacin [246]. In another study, it was found that the effects of norfloxacin on M. aeruginosa were modulated by light, the lowest EC50 being around 45 µg L−1 at high light intensity [249]. Ciprofloxacin was very toxic also to Microcystis panniformis, with an EC50 of 13.56 µg L−1 [171]. The EC50 values of gatifloxacin and moxifloxacin measured in Chlamydomonas reinhardtii, over 4–12 days of culturing, decreased progressively from 12.65 to 5.51 mg L−1 and from 83.04 to 17.39 mg L−1, respectively [261]. Their toxicity to M. aeruginosa was about three orders of magnitude greater, with 96 h EC50 of 25.30 and 60.34 µg L−1, respectively [248]. In summary, FQs are highly toxic to cyanobacteria since, in the majority of cases, their EC50 is lower than 1 mg L−1. This is not surprising as they are designed to act against prokaryotes. However, only a few classes of antibiotics (i.e., Aminoglycosides, Beta-lactams, Macrolides) have shown a comparable degree of toxicity toward cyanobacteria [280].

5.2. Unicellular Green Algae

Many authors have published data on the toxicity of FQs to Raphidocelis subcapitata, formerly known as Selenastrum capricornutum. In this green alga, Aderemi and co-authors evaluated the effects of ciprofloxacin and found a Lowest Observed Effect Concentration (LOEC) for growth inhibition of 6.329 mg L−1, whilst the most sensitive physiological endpoint among the various ones considered (superoxide dismutase activity, lipid peroxidation level, total energy content, energy consumption, and cellular energy allocation) was increased energy consumption (LOEC = 3.810 mg L−1), which was interpreted as the consequence of the need to respond to oxidative stress [281]. Lower concentrations of ciprofloxacin, corresponding to those measured in hospital wastewater (range 0.01–100 µg L−1), were assayed on R. subcapitata by Mater and colleagues [282], who found no effect on growth; however, when ciprofloxacin was mixed with analogous concentrations of tamoxifen or tamoxifen plus cyclophosphamide, a synergistic effect was evidenced, even at the lowest level assayed (0.01 µg L−1 of each compound). Liu and colleagues investigated the effects of ciprofloxacin on the photosynthetic process of R. subcapitata by determining various parameters, including photosynthetic rate, chlorophyll fluorescence, Hill Reaction, and ribulose-1.5-bisphosphate carboxylase activity [283]. They observed a significant reduction in the photosynthetic activity under concentrations ≥ 1 mg L−1. On the same green alga, Magdaleno et al. [250] assayed the growth inhibition activity of six compounds pertaining to different classes of antibacterials and found that ciprofloxacin was the most toxic, with an EC50 of 11.3 mg L−1, and an EC10 of 3.3 mg L−1. Once again, ciprofloxacin showed synergistic effects when assayed in binary mixtures (ratio 1:1) with gentamicin or vancomycin. The growth inhibition of ciprofloxacin in R. subcapitata was studied also in [246,251], who reported EC50s of 2.97 and 4.83 mg L−1, respectively. Robinson et al. assayed the toxicity of seven FQs on various aquatic organisms [245]; clinafloxacin was the most toxic to R. subcapitata, with an EC50 of 1.1 mg L−1. The toxicity of norfloxacin and levofloxacin to R. subcapitata was investigated by Gonzalez-Pleiter and colleagues [244]; they observed only moderate effects on algal growth at the highest assayed concentration; both norfloxacin (120 mg L−1) and levofloxacin (80 mg L−1) caused less than 50% inhibition. However, the two compounds showed synergistic interaction in binary mixtures when combined together or with other antibiotics such as erytromicin and tetracyclin. The results of Yamashita et al. regarding levofloxacin toxicity were quite different, with an EC50 of 1.2 mg L−1, an LOEC of 0.360 mg L−1, and a total inhibition of R. subcapitata growth at concentrations exceeding 2.5 mg L−1 [257]. For the same endpoint, Yang et al. reported an LOEC of 16 mg L−1 for norfloxacin and 5 mg L−1 for ciprofloxacin [252]. Eguchi et al. estimated norfloxacin EC50 values of 16.6 and 10.4 mg L−1 in the green algae R. subcapitata and Chlorella vulgaris, respectively [258]. Fu et al. evaluated the toxicity of three FQs to R. subcapitata [253]; their calculated growth inhibition EC50s after 96 h of exposure correspond to 7.082 (ciprofloxacin), 59.404 (norfloxacin), and 4.241 mg L−1 (ofloxacin). Indeed, the high toxicity of ofloxacin to green algae had been previously evidenced by Isidori et al. and Ferrari et al. with reported EC50s of 1.44 and 4.74 mg L−1, respectively [259,260]. Lützhøft et al. evaluated the toxicity to R. subcapitata of two FQs used in aquaculture, namely flumequine and sarafloxacin [247]; the reported EC50s for growth inhibition were 5 and 16 mg L−1, respectively. The toxicity of flumequine toward R. subcapitata was evaluated also by Zounková et al. [254], Van Der Grinten et al. [255], and Christensen et al. [256]; their calculated EC50s were in the range of 2.6–16 mg L−1.
Data regarding other algal organisms are also available. Backhaus et al. assayed various FQs on Scenedesmus vacuolatus [265]; flumequine was distinctly the most toxic, with an EC50 of 3.7 mg L−1. Andrieu et al. measured the toxicity of enrofloxacin and ciprofloxacin in Chlorella sp. [53]; the calculated EC50s were 111 and 23 mg L−1, respectively. Ciprofloxacin toxicity to C. vulgaris was investigated also by [262,263], who reported an EC50 of 29.09 and 20.6 mg L−1 for growth inhibition, respectively. Both enrofloxacin and ciprofloxacin were assayed on the freshwater green alga Desmodesmus subspicatus: calculated EC50s for growth inhibition were 5.568 and >8.042 mg L−1, respectively [243]. Enrofloxacin was assayed also on Scenedesmus obliquus, resulting in an EC50 of 45.10 mg L−1 after 72 h of exposure [266], whilst norfloxacin EC50 in the same species was 38.49 mg L−1 [267] or 50.18 mg L−1 [268]. Li and co-authors assayed moxifloxacin on Chlorella sorokiniana and Scenedesmus dimorphus [264]; their sensitivity was almost identical, with EC50s of 28 and 26 mg L−1, respectively. In summary, the EC50 of FQs in unicellular algae ranges from 1 to 100 mg L−1, confirming that the toxicity of antibiotics for unicellular algae may be orders of magnitude lower than for cyanobacteria. It is worth noting, however, that in P. subcapitata, which is the most extensively tested species, EC50s of less than 10 mg L−1 are found for many FQs.

5.3. Aquatic Plants

Among the different classes of antibiotics tested for phytotoxicity, FQs have generally proven to be the most potent class [284]. Ebert and colleagues evaluated the toxicity of enrofloxacin and ciprofloxacin on two freshwater macrophytes: the monocotyledonous Lemna minor and the dicotyledonous Myriophyllum spicatum [243]. L. minor showed to be far more sensitive to FQs than M. spicatum: EC50s of enrofloxacin and ciprofloxacin were 107 and 62.5 µg L−1, respectively. Similar results with these two compounds had already been published by Robinson et al., who also assayed the toxicity of five other FQs: levofloxacin, clinafloxacin, lomefloxacin, ofloxacin, and flumequine [245]. All these compounds showed themselves to be distinctly toxic to L. minor, their EC50s being in the range of 51–126 µg L−1, except for flumequine (EC50 2470 µg L−1). Flumequine effects on the aquatic weed Lythrum salicaria were investigated by Migliore et al. [285]: a significant growth inhibition was observed only at 100 mg L−1, whilst low concentrations (50–500 µg L−1) induced hormesis on secondary roots. Nunes and co-authors showed the activation of the anti-oxidant defensive system in both L. minor and Lemna gibba after exposure to very low concentrations (range 5–195 µg L−1) of ciprofloxacin [286].
A study on mesocosm wetlands planted with Phragmites australis [287] revealed browning and eventual die-off of the plant because of exposure to 2 mg L−1 of ciprofloxacin for a period of 5 days. Brain and colleagues evaluated the toxicity of four FQs in L. gibba; the calculated EC50s for wet mass reduction were the following: 97 µg L−1 of lomefloxacin, 185 µg L−1 of levofloxacin, 532 µg L−1 of ofloxacin, 698 µg L−1 of ciprofloxacin [269]. In the same species, the EC50 of norfloxacin was 913 µg L−1 [270]. In summary, the antibiotic FQs exhibit a toxicity to aquatic plants close to that observed for cyanobacteria and significantly higher than that exhibited towards unicellular algae. This is a crucial aspect of FQ ecotoxicity. Indeed, the presence and diversity of aquatic plants are essential for the health and functioning of aquatic ecosystems, providing numerous benefits to fish and crustaceans and other aquatic organisms by offering habitat, food, oxygen, refuge from predators, places for spawning and rearing young, and contributing to overall ecosystem stability and resilience.

5.4. Crustaceans

The toxicity of FQs to cladoceran crustaceans has been evaluated by various authors. Isidori and colleagues assayed the toxicity of ofloxacin in D. magna and Ceriodaphnia dubia. The latter was more sensitive to the antibacterial, with calculated EC50s of 17.41 mg L−1 (48 h, immobilization test) and 3.13 mg L−1 (7-day, population growth inhibition) [259]. Using the Comet assay, Nunes et al. evidenced genotoxic effects in D. magna exposed to ciprofloxacin at a concentration ≥ 0.013 mg L−1, whilst they observed no effects in the life-history parameters of the crustacean up to 0.195 mg L−1 [225]. Kergaravat and co-authors measured the Lethal Concentration, 50% (LC50), of various FQs both in D. magna and C. dubia [275]. The authors showed that after 72 h of exposure, toxicity is about 3–4 times higher than after 48 h, with LC50 values <10 mg L−1 for the most active compounds (i.e., levofloxacin and moxifloxacin). They also reported results of the chronic reproduction test with the same two cladocerans; again, levofloxacin and moxifloxacin were the most toxic, with No Observed Effect Concentration (NOEC) values in D. magna of <2.5 and <1.6 mg L−1, respectively. Mala and Dutta evaluated the toxicity of gemifloxacin and gatifloxacin in D. magna [274]. Their EC50s were two orders of magnitude greater than those of other FQs from previous generations, with values of 489.2 and 330.8 mg L−1, respectively. Eluk and co-authors evaluated both the 48 h EC50 and the 21-day NOEC of six FQs in D. magna [271]. The range of the estimated EC50 values was between 5.4 (marbofloxacin) and 36 mg L−1 (ofloxacin); the NOEC range was between 56 (ciprofloxacin) and 141 µg L−1 (ofloxacin). With regard to lomefloxacin, Luo et al. showed that exposure of D. magna under simulated sunlight radiation caused a remarkable increase in toxicity, with EC50 dropping from 166 to 64 mg L−1 [276]. Pan and co-authors evidenced effects on swimming activity and feeding rate in D. magna exposed to 25 mg L−1 of norfloxacin [288]. Dalla Bona and colleagues evaluated the acute toxicity of ciprofloxacin and enrofloxacin both in D. magna and Daphnia curvirostris: the latter showed to be more sensitive to the two FQs, with EC50s of 4.45 and 4.33 mg L−1, respectively [273]. In both species, binary mixtures of the two compounds showed mainly sub-additive interaction. In multigenerational tests conducted in D. magna, enrofloxacin [289] and flumequine [20] exhibited intergenerational and transgenerational toxicity, respectively. It is worth noting that the observed phenotypic changes in daphnids did not impact all individuals within the specific experimental group. Rather, they manifested only in certain subjects, following a stochastic pattern that could potentially indicate an interaction of the two FQs with the genetic material. In the same freshwater organism, enrofloxacin, flumequine, and levofloxacin also showed the ability to induce delayed toxicity after neonatal exposure, with EC50s recalculated after a follow-up of 10 days in pure medium, being 3.13, 7.18, and 15.11 mg L−1, respectively. Delayed toxicity was also observed after embryonic exposure of the crustacean to the three FQs, whilst their interaction in binary and ternary mixtures was merely additive [21]. Pietropoli and co-authors assessed the developmental and reproductive impairment caused by flumequine in D. magna, using an in vivo transcriptomic approach [19]. The lowest assayed concentration (0.2 mg L−1) had no noticeable impact on phenotypic traits; however, it did affect gene expression, and this effect became more pronounced at the highest exposure level (2 mg L−1), where phenotypic effects were evident and a significant modulation was detected in various genes associated with growth, development, structural components, and the antioxidant response. Dionisio and colleagues obtained an EC50 of 36.493 mg L−1 after acute exposure of D. magna to ciprofloxacin [272]; interestingly, they also showed that chronic exposure to relevant concentrations of ciprofloxacin may cause oxidative stress and that exposure of mothers to 3 mg L−1 of the compound may diminish the size of newborn daphnids. Ciprofloxacin and Enrofloxacin were assayed also on Moina macrocopa; their 48 h EC50s were 71.2 and 69.1 mg L−1, respectively [53]. Zhang and co-authors evaluated the toxic effects of enrofloxacin in the giant freshwater prawn Macrobrachium rosenbergii [290]. One-week exposure of juvenile individuals to 1–5 mg L−1 of the pharmaceutical caused dose-dependent growth inhibition, whilst a concentration of 0.2 mg L−1 had growth-promoting effects. Instead, no toxic effects were observed in Thamnocephalus platyurus exposed to 100 mg L−1 of levofloxacin [291]. In summary, the toxicity of FQs to freshwater crustaceans can be considered moderate, EC50s being usually in the order of tens of mg L−1; however, their evidenced ability to interact with gene expression and cause delayed and transgenerational effects suggests a possible impact on crustacean populations, even after short exposure in the natural environment, that the standard acute immobilization test cannot predict [19,20,21,273,289,292,293].

5.5. Fish

The toxicity of various FQs has been assayed on zebrafish (Danio rerio) embryos and, for all the compounds, crucial toxic effects on development were reported but only at concentrations of hundreds of mg L−1 [277]. Similar findings were reported by [242,278] after exposing zebrafish embryos to norfloxacin and enrofloxacin, respectively. Exposure of zebrafish to high concentrations of gatifloxacin reduced heart rate and cardiac output and resulted in pericardial edema [294]. In the same species, flumequine was shown to be teratogenic at relatively low concentrations (4.8–77 mg L−1), with an EC50 for embryo mortality of ~40 mg L−1 [279]. Nogueira and co-authors reported that 96 h exposure to ciprofloxacin (0.005–0.488 µg L−1) induced an increase in AChE (acetylcholinesterase) activity and a decrease in CAT (catalase) activity in D. rerio embryos or larvae [295]. Interestingly, 10 mg L−1 of norfloxacin caused a significant serum increase in vitellogenin in male goldfish (Carassius auratus). Moreover, in the same species, higher levels of norfloxacin were able to induce concentration- and time-dependent DNA alterations [296]. In Xiphophorus helleri, it has been shown that norfloxacin, even at a concentration < 1 mg L−1, can have effects on hepatic gene expression of P450 isoforms, GST (glutathione S-transferase), and P-glycoprotein [297]. Early-stage zebrafish exposed to norfloxacin nicotinate exhibited increased transcriptional levels and activities of major antioxidant enzymes like SOD (superoxide dismutase), CAT, and GPX (glutathione peroxidase) [298]. In juvenile common carp, Zhao and colleagues showed oxidative stress, damage to the intestinal barrier function, and change in the expression of immune-related genes after chronic exposure to very low concentrations of norfloxacin [299]. Yang and co-authors reported that ofloxacin, at concentrations > 0.05 mg L−1, significantly inhibits AChE, promotes EROD (7-ethoxyresorufin-O-deethylase), and increases SOD activities in C. auratus [300]. In the fathead minnow (Pimephales promelas, 7-day early life stage) seven FQs (i.e., ciprofloxacin, clinafloxacin, enrofloxacin, flumequine, levofloxacin, lomefloxacin, and ofloxacin) have been tested for effects on survival and growth [245]. The compounds showed limited toxicity, with NOEC at 10 mg L−1, the only exception being clinafloxacin, with 100% lethality at 10 mg L−1 (NOEC = 2 mg L−1). Enrofloxacin (11 mg L−1) caused 80% mortality in medaka fish (Orizia latipes) after 30-day continuous post-hatching exposure [301]. However, acute exposure (96 h) up to 100 mg L−1 caused no effects in juveniles (10–14 days post hatch) of the same species [302]. Likewise, acute post-hatching exposure (96 h) to 100 mg L−1 levofloxacin did not cause any acute toxic effect [291]. Zhang and colleagues evaluated the toxicity of an equi-concentrations mixture of four FQs (i.e., ciprofloxacin, ofloxacin, norfloxacin, enrofloxacin) on D. rerio larvae and estimated an EC50 of 481.3 mg L−1, based on a malformation rate endpoint [303]. In summary, fish have been shown to be less sensitive than other aquatic organisms to the acute toxicity of FQs. However, the ability of FQs to interact with gene expression, interfere with embryo development, and increase serum vitellogenin once again suggests a possible impact on populations, even after short exposure in the natural environment.

5.6. Other Freshwater Organisms

Very few data are available concerning the toxicity of FQs towards other freshwater organisms. He and colleagues [304] evaluated the toxicity of norfloxacin and ofloxacin in young Bellamya aeruginosa snail. At concentrations up to 300 mg L−1, no lethal effects were observed. However, they obtained EC50s values based on activity inhibition rate. This effect was concentration- and exposure-time-dependent. After 48 h exposure, the calculated EC50s were 141.3 (norfloxacin) and 222.6 (ofloxacin) mg L−1. The scarce toxicity (EC50>100 mg L−1) to mollusks was confirmed as well with moxifloxacin in the bivalve Lampsilis siliquoidea [305]. A reproduction study exposing the snail Potamopyrgus antipodarum for 52 days to 0.8 μg L−1 ciprofloxacin elicited no effects [306]. Overall, and considering also data obtained on marine mollusks, almost 75% of the tested FQs demonstrated negligible toxicity to this taxon [24].
A sediment toxicity study investigated reproduction effects of ciprofloxacin in Lumbriculus variegatus and Chironomus riparius during 28 days of exposure. Both species were exposed to 0.25, 0.5, 1.0, 2.0, and 4.0 μg kg−1, which did not cause any significant effects [306].
Ciprofloxacin and levofloxacin were assayed on Xenopus laevis larvae [307], but the highest concentration of the two FQs (100 mg L−1) showed no effects on body length after 96 h exposure.

6. Risk Assessment

Overall, data indicate for freshwater organisms the following rank of sensitivity to FQs: cyanobacteria > aquatic plants > unicellular algae > crustaceans > fish. In Figure 2, to give an overview of the susceptibility of the aquatic biota to the toxicity of the various FQs, EC50 values obtained in the various taxa are presented.
When data are available for a reasonable number of species, a risk evaluation can be performed using the methodology outlined in [308]. Accordingly, results from eleven distinct FQ compounds across twelve diverse autotroph species were amalgamated in a Species Sensitivity Distribution (SSD) curve. By normalizing the acute toxicity values (EC50s) of each FQ to its EC50 in M. aeruginosa, the freshwater cyanobacterium highly sensitive to all FQs tested, the EC50s were expressed as Microcystis-equivalents, enabling cross-FQs pooling. The set of geometric mean Microcystis-equivalent EC50s (Table 2) was then analyzed using the USEPA SSD Generator V1, available at [309]. The endpoints considered to build the nSSD were: growth/yield inhibition (48 or 72 h) for cyanobacteria and unicellular algae, and seven-day growth/yield inhibition for plants. The decision to limit the nSSD curve to autotrophic species is motivated by the fact that they are more sensitive than heterotrophs to FQs and that, consequently, a safety threshold calculated for them can be a guarantee for all freshwater species.
In Figure 3, the Microcystis-equivalent SSD encompassing twelve species of autotroph organisms is illustrated, with prediction intervals and data points that led to the acquisition of the fifth percentile (Hazard Concentration, HC5), corresponding to 0.815, with a 95% confidence interval of 0.143–4.653. Multiplying this normalized value by the EC50 measured for each FQ in M. aeruginosa, the following individual HC5 values were then obtained: 7.51 µg L−1 (ciprofloxacin); 12.23 µg L−1 (clinafloxacin); 39.94 µg L−1 (enrofloxacin); 454.97 µg L−1 (flumequine); 20.62 µg L−1 (gatifloxacin); 6.52 µg L−1 (levofloxacin); 151.59 µg L−1 (lomefloxacin); 49.18 µg L−1 (moxifloxacin); 36.68 µg L−1 (norfloxacin); 17.12 µg L−1 (ofloxacin); 12.23 µg L−1 (sarafloxacin). These HC5 values may serve as effective metrics for preliminary risk assessments, as the comprehensive SSD can be harmonized with estimated exposure distributions for more comprehensive risk characterization [308].
The normalized SSD for FQs provides a more comprehensive and statistically robust foundation for risk assessment in comparison to data from a single FQ in a single most sensitive species, which is instead advantageous for assessing compounds with data available for a limited number of species. Present-day risk assessment techniques incorporate an Assessment Factor (AF) that is adapted according to the data employed in SSD development and subsequently applied to the HC5 [311]. Ordinarily, it is recommended that the HC5 value be estimated from toxicity data including at least 4–8 species, and it is divided by a scaling AF ranging from 1 to 5 to determine the PNEC [312,313]. Even applying the most conservative AF of 5, the PNEC values calculated for the various FQs are higher, by about one order of magnitude, when compared to those obtained using an AF of 100 applied to the lowest EC50 in cyanobacteria (Figure 4). It is important to emphasize that the substantial AF (100) in the standard algorithm for single-compound risk evaluation primarily addresses the risk of overstating the PNEC in the face of extensive species sensitivity variations. Conversely, PNEC estimation through the SSD approach hinges predominantly on the number of species considered [312]. In the present analysis, twelve different species from the most sensitive taxa were included, suggesting an acceptable accuracy in PNEC determination, further supported by the high R2 value of the SSD curve (0.912).
Constructing an analogous SSD for heterotrophic organisms was hindered by data limitations, particularly concerning the absence of data for taxa such as mollusks and insects, along with sparse information available for amphibians and fish. The scarcity of data on fish results from their relatively lower sensitivity to FQs, leading to their infrequent use in acute toxicity tests. Moreover, the few available EC50 data for fish often fall into the “greater than” range, as it is not recommended to test concentrations above 100 mg L−1. Indeed, this concentration threshold is defined as a “limit test” by OECD Guideline 203 on “acute fish toxicity test” [314].
To allow for risk evaluation, in Figure 4, the PNEC, obtained either by applying an assessment factor (100 or 1000) to the lowest EC50 of each compound in the various freshwater taxa [315] or an assessment factor of 5 to the Hazard Concentration fifth percentile, is compared with the box plot of the highest freshwater concentration reported in the literature.
For the majority of compounds, the highest reported concentration is higher than the PNEC obtained by applying an assessment factor of 100 to the lowest EC50 in cyanobacteria. This is not surprising as the level of contamination in some specific areas of some countries in the Far East is extraordinarily high, mainly due to the activities of pharmaceutical companies and fish farms. Interestingly, with four FQs largely used in human medicine, namely ciprofloxacin, norfloxacin, ofloxacin, and sarafloxacin, even the median of the highest concentration detected in the aquatic environment is higher than the PNEC calculated in the traditional way; moreover, for two additional human pharmaceuticals (moxifloxacin and levofloxacin), the median is less than one order of magnitude lower than the PNEC. With regard to veterinary compounds, enrofloxacin, with a PNEC only an order of magnitude lower than the median, is still widely used in food-producing animals and could pose a risk to the aquatic environment in the years to come, particularly in countries where its use is also permitted in aquaculture. Notably, according to [316], six of the eleven largest aquaculture-producing countries currently use enrofloxacin.
With reference to the PNECs obtained from the nSSD curve, that, as we said, are one order of magnitude higher, there are still five FQs (ciprofloxacin, enrofloxacin, norfloxacin, ofloxacin, and sarafloxacin) with reported concentrations in the freshwater environment above the threshold. Consequently, a risk to the aquatic environment cannot be ruled out, particularly in those countries (India, China) where very high concentrations were most often detected. It should be noted, however, that an enormous effort has been made in China during the last decade in the search for drugs in the freshwater environment, and, as a consequence, a worse picture may have been obtained than in other countries. The risk regards autotroph organisms in particular and, considering their fundamental role in sustaining life of higher trophic level, bottom-up repercussions are inevitable. Indeed, they provide the aquatic fauna with not only food but also habitat, oxygen, and nutrient cycling. Macrophytes, in particular, can improve water quality by absorbing excess nutrients and pollutants, help stabilize sediments, provide refuge from predators and places for spawning and rearing young.
In a recent review from Thai and colleagues [25], regarding quinolone antibiotics in the Baiyangdian Lake (China), PNECs for flumequine and ofloxacin were presented, based on SSDs that included both autotrophic and heterotrophic species. They applied, like us, an AF of 5 and obtained PNEC values of 4.40 and 196 µg L−1 for ofloxacin and flumequine, respectively. These values are higher than those obtained by us (3.42 and 91 µg L−1). Of course, these differences could be justified by the exclusive use of autotroph organisms, the most sensitive taxa, to construct our nSSD curve.
Risk assessment should also consider the possible active environmental metabolites of FQs. These may be the result of enzymatic activity in the human [317] and the animal body [318,319] or the product of biotransformation by bacteria and fungi [320]. Photolysis products can also be active and, in some cases, even more toxic than the parent compound [321]. Unfortunately, comprehensive knowledge of all possible active metabolites of the various FQs is far from being achieved, partly because of the sophisticated techniques required for their identification and the limited availability of specific analytical standards for performing ecotoxicity assays.
Another aspect that should be considered in risk evaluation is the possible simultaneous presence of various FQs in a contaminated aquatic ecosystem. For mixtures of compounds having the same mechanism of action (MoA), the reference gold standard for predicting toxicological interaction is the Concentration Addition principle [322]. Accordingly, each chemical contribution to the overall toxicity of a mixture can be expressed as the quotient of its dose in the mixture and the dose of the same chemical alone that would be required to elicit the effect of the whole mixture. The few experiments carried out so far, on daphnids [21,273], indicate that binary and ternary mixtures of FQs display either additive or less-than-additive interaction. A similar conclusion was drawn by Backhaus et al. after assaying the toxicity of quinolone mixtures on Vibrio fischeri [323]. These findings are generally consistent with the data generated so far on the toxicity of chemical mixtures, as no interaction (simple additivity) or antagonism (less than additivity) has been observed in 95% of cases [324]. However, caution must be exercised when applying these results to the entire class of FQs: the fact that FQs share the same MoA does not necessarily imply that all potential combinations will not display synergistic interactions. For instance, Khadra et al. demonstrated a synergistic interaction in a ternary mixture composed of two FQs (ciprofloxacin and enrofloxacin) and one quinolone (nalidixic acid) when evaluating its genotoxicity in Vicia faba [325].
Overall, as autotrophs are far more sensitive than heterotrophs to FQ toxicity, a general safety threshold for FQs in freshwater could be assumed based on the HC5 dataset derived from the normalized SSD curve of autotrophs alone. Applying the most conservative AF (i.e., 5) to the lowest individual HC5 (6.52 µg L−1, levofloxacin), this safety threshold could be tentatively set at 1.30 µg L−1 (95% CI 0.23–7.44 µg L−1). Nonetheless, it would be advisable to determine such a threshold as the aggregate of all potential active ingredients stemming from FQ contamination within the freshwater ecosystem, metabolites included. Indeed, given that even low concentrations of single compounds may lead to severe overall toxicity when acting simultaneously on an organism, the toxicity of mixtures of pollutants has to be considered for the setting of environmental standards [323].
Finally, there is a lack of data on the toxicity of FQs to benthic fauna. This remains an important gap to fill, considering the tendency of FQs to accumulate in sediments. Indeed, the risk assessment presented here is based on the concentrations reported in the water column, which are generally one thousand times lower than those found in sediments [30]. In this regard, it should be emphasized that any damage to the biodiversity of freshwater benthic fauna can hinder the nutrient cycle that sustains life at all levels. Of course, benthic invertebrates play a crucial role by breaking down the organic detritus, mixing and aerating sediments, and promoting microbial growth [326].

7. Conclusions

Although FQs are recognized worldwide as “critically important antimicrobials”, they have been used indiscriminately in both human and veterinary health care in recent decades. Their limited metabolization and relative resistance to degradation have led to their widespread presence in municipal wastewater, as well as in manure/slurry, and to their consequent occurrence in various environmental compartments, including surface waters. In some specific areas, pharmaceutical production plants and aquaculture facilities have also contributed significantly to the contamination of watersheds. The short-term toxicity of FQs to freshwater organisms is remarkable, with EC50s lower than 10 µg L−1 in cyanobacteria and 100 µg L−1 in aquatic plants. Indeed, for five FQs (ciprofloxacin, enrofloxacin, norfloxacin, ofloxacin, and sarafloxacin), the highest concentrations detected in the environment, when matched to the PNEC, give a Risk Quotient > 1. Moreover, FQs may induce delayed toxic effects and express embryonic and transgenerational toxicity in crustaceans: all features that are presumably related to their ability to interact with the genome of non-target organisms and may add complexity to risk assessment. Even in fish, which are far less sensitive than other aquatic organisms to the toxicity of FQs, toxic effects have been reported (e.g., genotoxicity, ECM degradation, neurotoxicity) and their putative underlying mechanisms postulated. Generally, our knowledge regarding the mechanisms of action in non-target species within freshwater environments is limited. Nevertheless, the existing information is already cause for concern.
More specifically, the use of this class of antimicrobials should be limited for many reasons:
  • Their particular efficacy in treating a range of dangerous bacterial infections should be safeguarded, limiting the spread of drug resistance as much as possible;
  • As with any class of antibiotics, limiting their use helps to maintain the ecological integrity of aquatic microbial communities;
  • The extent of their removal by conventional wastewater treatment plants is unsatisfactory;
  • Contamination of the freshwater environment is already widespread, and, in some cases, they achieve remarkably high levels;
  • Their relative resistance to degradation combined with a strong tendency to adsorption favors their progressive accumulation in sediments;
  • Their pronounced toxicity to organisms at basal trophic levels can have bottom-up repercussions in aquatic ecosystems;
  • Their potential ability to disrupt aquatic plant communities may influence the structure of the habitats provided by plants to various organisms;
  • Little is known about possible toxicological interactions amongst FQs or with other co-occurring contaminants of the freshwater environment;
  • Their possible transgenerational and delayed toxicity increases the complexity of defining environmental safety thresholds;
  • Given their ability to interact with the genome, even brief occasional exposure of freshwater organisms to FQs could have long-term consequences;
  • The restriction of FQ use aligns with broader efforts to promote sustainable water management practices, emphasizing the protection and conservation of aquatic ecosystems for future generations.
Overall, the toxicity to freshwater organisms of the eleven compounds considered here does not appear to vary significantly among FQ generations (Figure 2); however, there are at least eight newer FQs on the market [3], not mentioned here, for which data on both ecotoxicity and presence in the freshwater environment are not yet available. It is hoped that data will also be collected for these more modern FQs in the coming years, to verify whether their environmental impact is lower than that of their predecessors. It will also be essential to perform ecotoxicity tests on different benthic organisms to better assess the risk that FQs pose to the aquatic environment; in this context, their ability to cause delayed, multigenerational, and transgenerational toxic effects in crustaceans should not be overlooked. Finally, future research should also focus on new cost-effective techniques to improve the efficiency of sewage treatment plants in removing FQs.

Author Contributions

Conceptualization, M.D.L. and M.P.; formal analysis, M.D.L.; investigation, M.D.L. and M.P.; writing—original draft preparation, M.D.L. and M.P.; funding acquisition, M.D.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the University of Padua, SID 2020 grant (Prot. BIRD 201185).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

The authors are grateful to Jonathan Dancer (MRPharmS) for contributing to the language used in the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Emmerson, A.M. The Quinolones: Decades of Development and Use. J. Antimicrob. Chemother. 2003, 51, 13–20. [Google Scholar] [CrossRef] [PubMed]
  2. Marchant, J. When Antibiotics Turn Toxic. Nature 2018, 555, 431–433. [Google Scholar] [CrossRef] [PubMed]
  3. Rusu, A.; Munteanu, A.-C.; Arbănași, E.-M.; Uivarosi, V. Overview of Side-Effects of Antibacterial Fluoroquinolones: New Drugs versus Old Drugs, a Step Forward in the Safety Profile? Pharmaceutics 2023, 15, 804. [Google Scholar] [CrossRef] [PubMed]
  4. Idowu, T.; Schweizer, F. Ubiquitous Nature of Fluoroquinolones: The Oscillation between Antibacterial and Anticancer Activities. Antibiotics 2017, 6, 26. [Google Scholar] [CrossRef] [PubMed]
  5. Martinez, M.; McDermott, P.; Walker, R. Pharmacology of the Fluoroquinolones: A Perspective for the Use in Domestic Animals. Vet. J. 2006, 172, 10–28. [Google Scholar] [CrossRef] [PubMed]
  6. Schmerold, I.; Van Geijlswijk, I.; Gehring, R. European Regulations on the Use of Antibiotics in Veterinary Medicine. Eur. J. Pharm. Sci. 2023, 189, 106473. [Google Scholar] [CrossRef] [PubMed]
  7. Blondeau, J.M. Fluoroquinolones: Mechanism of Action, Classification, and Development of Resistance. Surv. Ophthalmol. 2004, 49, S73–S78. [Google Scholar] [CrossRef] [PubMed]
  8. Thompson, R.J.; Mosig, G. An ATP-Dependent Supercoiling Topoisomerase of Chlamydomonas reinhardtii Affects Accumulation of Specific Chforoplast Transcripts. Nucleic Acids Res. 1985, 13, 873–891. [Google Scholar] [CrossRef] [PubMed]
  9. Wall, M.K.; Mitchenall, L.A.; Maxwell, A. Arabidopsis thaliana DNA Gyrase Is Targeted to Chloroplasts and Mitochondria. Proc. Natl. Acad. Sci. USA 2004, 101, 7821–7826. [Google Scholar] [CrossRef]
  10. Hangas, A.; Aasumets, K.; Kekäläinen, N.J.; Paloheinä, M.; Pohjoismäki, J.L.; Gerhold, J.M.; Goffart, S. Ciprofloxacin Impairs Mitochondrial DNA Replication Initiation through Inhibition of Topoisomerase 2. Nucleic Acids Res. 2018, 46, 9625–9636. [Google Scholar] [CrossRef]
  11. Mercer, M.A. Quinolones, Including Fluoroquinolones—Use in Animals 2022. Available online: https://www.msdvetmanual.com/pharmacology/antibacterial-agents/quinolones,-including-fluoroquinolones,-use-in-animals (accessed on 15 May 2024).
  12. Badal, S.; Her, Y.F.; Maher, L.J. Nonantibiotic Effects of Fluoroquinolones in Mammalian Cells. J. Biol. Chem. 2015, 290, 22287–22297. [Google Scholar] [CrossRef] [PubMed]
  13. Tennyson, L.E.; Averch, T.D. An Update on Fluoroquinolones: The Emergence of a Multisystem Toxicity Syndrome. Urol. Pract. 2017, 4, 383–387. [Google Scholar] [CrossRef] [PubMed]
  14. European Medicines Agency. Fluoroquinolone and Quinolone Antibiotics: PRAC Recommends New Restrictions on Use; European Medicines Agency: Amsterdam, The Netherlands, 2018. [Google Scholar]
  15. Ly, N.F.; Flach, C.; Lysen, T.S.; Markov, E.; Van Ballegooijen, H.; Rijnbeek, P.; Duarte-Salles, T.; Reyes, C.; John, L.H.; Karimi, L.; et al. Impact of European Union Label Changes for Fluoroquinolone-Containing Medicinal Products for Systemic and Inhalation Use: Post-Referral Prescribing Trends. Drug Saf. 2023, 46, 405–416. [Google Scholar] [CrossRef] [PubMed]
  16. Sukul, P.; Spiteller, M. Fluoroquinolone Antibiotics in the Environment. In Reviews of Environmental Contamination and Toxicology; Springer: New York, NY, USA, 2007; Volume 191, pp. 131–162. ISBN 978-0-387-69162-6. [Google Scholar]
  17. Riaz, L.; Mahmood, T.; Khalid, A.; Rashid, A.; Ahmed Siddique, M.B.; Kamal, A.; Coyne, M.S. Fluoroquinolones (FQs) in the Environment: A Review on Their Abundance, Sorption and Toxicity in Soil. Chemosphere 2018, 191, 704–720. [Google Scholar] [CrossRef] [PubMed]
  18. Välitalo, P.; Kruglova, A.; Mikola, A.; Vahala, R. Toxicological Impacts of Antibiotics on Aquatic Micro-Organisms: A Mini-Review. Int. J. Hyg. Environ. Health 2017, 220, 558–569. [Google Scholar] [CrossRef] [PubMed]
  19. Pietropoli, E.; Pauletto, M.; Tolosi, R.; Iori, S.; Lopparelli, R.M.; Montanucci, L.; Giantin, M.; Dacasto, M.; De Liguoro, M. An In Vivo Whole-Transcriptomic Approach to Assess Developmental and Reproductive Impairments Caused by Flumequine in Daphnia magna. Int. J. Mol. Sci. 2023, 24, 9396. [Google Scholar] [CrossRef] [PubMed]
  20. De Liguoro, M.; Maraj, S.; Merlanti, R. Transgenerational Toxicity of Flumequine over Four Generations of Daphnia magna. Ecotoxicol. Environ. Saf. 2019, 169, 814–821. [Google Scholar] [CrossRef] [PubMed]
  21. Tolosi, R.; De Liguoro, M. Delayed Toxicity of Three Fluoroquinolones and Their Mixtures after Neonatal or Embryonic Exposure, in Daphnia magna. Ecotoxicol. Environ. Saf. 2021, 225, 112778. [Google Scholar] [CrossRef] [PubMed]
  22. German Environment Agency. Database—Pharmaceuticals in the Environment; Umweltbundesamt—UBA: Dessau-Roßlau, Germany, 2020. [Google Scholar]
  23. Olker, J.H.; Elonen, C.M.; Pilli, A.; Anderson, A.; Kinziger, B.; Erickson, S.; Skopinski, M.; Pomplun, A.; LaLone, C.A.; Russom, C.L.; et al. The ECOTOXicology Knowledgebase: A Curated Database of Ecologically Relevant Toxicity Tests to Support Environmental Research and Risk Assessment. Environ. Toxicol. Chem. 2022, 41, 1520–1539. [Google Scholar] [CrossRef] [PubMed]
  24. Shen, M.; Hu, Y.; Zhao, K.; Li, C.; Liu, B.; Li, M.; Lyu, C.; Sun, L.; Zhong, S. Occurrence, Bioaccumulation, Metabolism and Ecotoxicity of Fluoroquinolones in the Aquatic Environment: A Review. Toxics 2023, 11, 966. [Google Scholar] [CrossRef]
  25. Zhang, L.; Shen, L.; Qin, S.; Cui, J.; Liu, Y. Quinolones Antibiotics in the Baiyangdian Lake, China: Occurrence, Distribution, Predicted No-Effect Concentrations (PNECs) and Ecological Risks by Three Methods. Environ. Pollut. 2020, 256, 113458. [Google Scholar] [CrossRef] [PubMed]
  26. Du, J.; Liu, Q.; Pan, Y.; Xu, S.; Li, H.; Tang, J. The Research Status, Potential Hazards and Toxicological Mechanisms of Fluoroquinolone Antibiotics in the Environment. Antibiotics 2023, 12, 1058. [Google Scholar] [CrossRef] [PubMed]
  27. Thai, V.-A.; Dang, V.D.; Thuy, N.T.; Pandit, B.; Vo, T.-K.-Q.; Khedulkar, A.P. Fluoroquinolones: Fate, Effects on the Environment and Selected Removal Methods. J. Clean. Prod. 2023, 418, 137762. [Google Scholar] [CrossRef]
  28. Bhatt, S.; Chatterjee, S. Fluoroquinolone Antibiotics: Occurrence, Mode of Action, Resistance, Environmental Detection, and Remediation—A Comprehensive Review. Environ. Pollut. 2022, 315, 120440. [Google Scholar] [CrossRef]
  29. Teglia, C.M.; Perez, F.A.; Michlig, N.; Repetti, M.R.; Goicoechea, H.C.; Culzoni, M.J. Occurrence, Distribution, and Ecological Risk of Fluoroquinolones in Rivers and Wastewaters. Environ. Toxicol. Chem. 2019, 38, 2305–2313. [Google Scholar] [CrossRef] [PubMed]
  30. Van Doorslaer, X.; Dewulf, J.; Van Langenhove, H.; Demeestere, K. Fluoroquinolone Antibiotics: An Emerging Class of Environmental Micropollutants. Sci. Total Environ. 2014, 500–501, 250–269. [Google Scholar] [CrossRef]
  31. Rusu, A.; Lungu, I.-A.; Moldovan, O.-L.; Tanase, C.; Hancu, G. Structural Characterization of the Millennial Antibacterial (Fluoro)Quinolones—Shaping the Fifth Generation. Pharmaceutics 2021, 13, 1289. [Google Scholar] [CrossRef]
  32. Mathur, P.; Sanyal, D.; Callahan, D.L.; Conlan, X.A.; Pfeffer, F.M. Treatment Technologies to Mitigate the Harmful Effects of Recalcitrant Fluoroquinolone Antibiotics on the Environment and Human Health. Environ. Pollut. 2021, 291, 118233. [Google Scholar] [CrossRef]
  33. Guo, X.; Yan, Z.; Zhang, Y.; Kong, X.; Kong, D.; Shan, Z.; Wang, N. Removal Mechanisms for Extremely High-Level Fluoroquinolone Antibiotics in Pharmaceutical Wastewater Treatment Plants. Environ. Sci. Pollut. Res. 2017, 24, 8769–8777. [Google Scholar] [CrossRef]
  34. Parente, C.E.T.; Azeredo, A.; Vollú, R.E.; Zonta, E.; Azevedo-Silva, C.E.; Brito, E.M.S.; Seldin, L.; Torres, J.P.M.; Meire, R.O.; Malm, O. Fluoroquinolones in Agricultural Soils: Multi-Temporal Variation and Risks in Rio de Janeiro Upland Region. Chemosphere 2019, 219, 409–417. [Google Scholar] [CrossRef]
  35. Parente, C.E.T.; Brito, E.M.S.; Azeredo, A.; Meire, R.O.; Malm, O. Fluoroquinolone Antibiotics and Their Interactions in Agricultural Soils—A Review. Orbital Electron. J. Chem. 2019, 11, 42–52. [Google Scholar] [CrossRef]
  36. Nowara, A.; Burhenne, J.; Spiteller, M. Binding of Fluoroquinolone Carboxylic Acid Derivatives to Clay Minerals. J. Agric. Food Chem. 1997, 45, 1459–1463. [Google Scholar] [CrossRef]
  37. Franklin, A.M.; Williams, C.; Andrews, D.M.; Watson, J.E. Sorption and Desorption Behavior of Four Antibiotics at Concentrations Simulating Wastewater Reuse in Agricultural and Forested Soils. Chemosphere 2022, 289, 133038. [Google Scholar] [CrossRef] [PubMed]
  38. Maghsodian, Z.; Sanati, A.M.; Mashifana, T.; Sillanpää, M.; Feng, S.; Nhat, T.; Ramavandi, B. Occurrence and Distribution of Antibiotics in the Water, Sediment, and Biota of Freshwater and Marine Environments: A Review. Antibiotics 2022, 11, 1461. [Google Scholar] [CrossRef]
  39. Wu, R.; Ruan, Y.; Huang, G.; Li, J.; Lao, J.-Y.; Lin, H.; Liu, Y.; Cui, Y.; Zhang, K.; Wang, Q.; et al. Source Apportionment, Hydrodynamic Influence, and Environmental Stress of Pharmaceuticals in a Microtidal Estuary with Multiple Outlets in South China. Environ. Sci. Technol. 2022, 56, 11374–11386. [Google Scholar] [CrossRef] [PubMed]
  40. Golet, E.M.; Alder, A.C.; Giger, W. Environmental Exposure and Risk Assessment of Fluoroquinolone Antibacterial Agents in Wastewater and River Water of the Glatt Valley Watershed, Switzerland. Environ. Sci. Technol. 2002, 36, 3645–3651. [Google Scholar] [CrossRef] [PubMed]
  41. Huang, L.; Mo, Y.; Wu, Z.; Rad, S.; Song, X.; Zeng, H.; Bashir, S.; Kang, B.; Chen, Z. Occurrence, Distribution, and Health Risk Assessment of Quinolone Antibiotics in Water, Sediment, and Fish Species of Qingshitan Reservoir, South China. Sci. Rep. 2020, 10, 15777. [Google Scholar] [CrossRef]
  42. Fick, J.; Söderström, H.; Lindberg, R.H.; Phan, C.; Tysklind, M.; Larsson, D.G.J. Contamination of Surface, Ground, and Drinking Water from Pharmaceutical Production. Environ. Toxicol. Chem. 2009, 28, 2522–2527. [Google Scholar] [CrossRef] [PubMed]
  43. Gothwal, R. Shashidhar Occurrence of High Levels of Fluoroquinolones in Aquatic Environment Due to Effluent Discharges from Bulk Drug Manufacturers. J. Hazard. Toxic Radioact. Waste 2017, 21, 05016003. [Google Scholar] [CrossRef]
  44. Larsson, D.G.J.; De Pedro, C.; Paxeus, N. Effluent from Drug Manufactures Contains Extremely High Levels of Pharmaceuticals. J. Hazard. Mater. 2007, 148, 751–755. [Google Scholar] [CrossRef]
  45. Le, T.X.; Munekage, Y. Residues of Selected Antibiotics in Water and Mud from Shrimp Ponds in Mangrove Areas in Viet Nam. Mar. Pollut. Bull. 2004, 49, 922–929. [Google Scholar] [CrossRef] [PubMed]
  46. López-Serna, R.; Pérez, S.; Ginebreda, A.; Petrović, M.; Barceló, D. Fully Automated Determination of 74 Pharmaceuticals in Environmental and Waste Waters by Online Solid Phase Extraction–Liquid Chromatography-Electrospray-Tandem Mass Spectrometry. Talanta 2010, 83, 410–424. [Google Scholar] [CrossRef] [PubMed]
  47. Yiruhan; Wang, Q.-J.; Mo, C.-H.; Li, Y.-W.; Gao, P.; Tai, Y.-P.; Zhang, Y.; Ruan, Z.-L.; Xu, J.-W. Determination of Four Fluoroquinolone Antibiotics in Tap Water in Guangzhou and Macao. Environ. Pollut. 2010, 158, 2350–2358. [Google Scholar] [CrossRef] [PubMed]
  48. Gao, L.; Shi, Y.; Li, W.; Liu, J.; Cai, Y. Occurrence, Distribution and Bioaccumulation of Antibiotics in the Haihe River in China. J. Environ. Monit. 2012, 14, 1248. [Google Scholar] [CrossRef] [PubMed]
  49. Al Aukidy, M.; Verlicchi, P.; Jelic, A.; Petrovic, M.; Barcelò, D. Monitoring Release of Pharmaceutical Compounds: Occurrence and Environmental Risk Assessment of Two WWTP Effluents and Their Receiving Bodies in the Po Valley, Italy. Sci. Total Environ. 2012, 438, 15–25. [Google Scholar] [CrossRef]
  50. Alexy, R.; Sommer, A.; Lange, F.T.; Kümmerer, K. Local Use of Antibiotics and Their Input and Fate in a Small Sewage Treatment Plant—Significance of Balancing and Analysis on a Local Scale vs. Nationwide Scale. Acta Hydrochim. Hydrobiol. 2006, 34, 587–592. [Google Scholar] [CrossRef]
  51. Alygizakis, N.A.; Besselink, H.; Paulus, G.K.; Oswald, P.; Hornstra, L.M.; Oswaldova, M.; Medema, G.; Thomaidis, N.S.; Behnisch, P.A.; Slobodnik, J. Characterization of Wastewater Effluents in the Danube River Basin with Chemical Screening, in Vitro Bioassays and Antibiotic Resistant Genes Analysis. Environ. Int. 2019, 127, 420–429. [Google Scholar] [CrossRef] [PubMed]
  52. Andreozzi, R.; Raffaele, M.; Nicklas, P. Pharmaceuticals in STP Effluents and Their Solar Photodegradation in Aquatic Environment. Chemosphere 2003, 50, 1319–1330. [Google Scholar] [CrossRef] [PubMed]
  53. Andrieu, M.; Rico, A.; Phu, T.M.; Huong, D.T.T.; Phuong, N.T.; Van Den Brink, P.J. Ecological Risk Assessment of the Antibiotic Enrofloxacin Applied to Pangasius Catfish Farms in the Mekong Delta, Vietnam. Chemosphere 2015, 119, 407–414. [Google Scholar] [CrossRef]
  54. Archana, G.; Dhodapkar, R.; Kumar, A. Offline Solid-Phase Extraction for Preconcentration of Pharmaceuticals and Personal Care Products in Environmental Water and Their Simultaneous Determination Using the Reversed Phase High-Performance Liquid Chromatography Method. Environ. Monit. Assess. 2016, 188, 512. [Google Scholar] [CrossRef]
  55. Archana, G.; Dhodapkar, R.; Kumar, A. Ecotoxicological Risk Assessment and Seasonal Variation of Some Pharmaceuticals and Personal Care Products in the Sewage Treatment Plant and Surface Water Bodies (Lakes). Environ. Monit. Assess. 2017, 189, 446. [Google Scholar] [CrossRef]
  56. Aydin, S.; Aydin, M.E.; Ulvi, A.; Kilic, H. Antibiotics in Hospital Effluents: Occurrence, Contribution to Urban Wastewater, Removal in a Wastewater Treatment Plant, and Environmental Risk Assessment. Environ. Sci. Pollut. Res. 2019, 26, 544–558. [Google Scholar] [CrossRef]
  57. Azuma, T.; Ishiuchi, H.; Inoyama, T.; Teranishi, Y.; Yamaoka, M.; Sato, T.; Mino, Y. Occurrence and Fate of Selected Anticancer, Antimicrobial, and Psychotropic Pharmaceuticals in an Urban River in a Subcatchment of the Yodo River Basin, Japan. Environ. Sci. Pollut. Res. 2015, 22, 18676–18686. [Google Scholar] [CrossRef]
  58. Bai, Y.; Meng, W.; Xu, J.; Zhang, Y.; Guo, C. Occurrence, Distribution and Bioaccumulation of Antibiotics in the Liao River Basin in China. Environ. Sci. Process. Impacts 2014, 16, 586. [Google Scholar] [CrossRef]
  59. Batt, A.L.; Aga, D.S. Simultaneous Analysis of Multiple Classes of Antibiotics by Ion Trap LC/MS/MS for Assessing Surface Water and Groundwater Contamination. Anal. Chem. 2005, 77, 2940–2947. [Google Scholar] [CrossRef]
  60. Batt, A.L.; Bruce, I.B.; Aga, D.S. Evaluating the Vulnerability of Surface Waters to Antibiotic Contamination from Varying Wastewater Treatment Plant Discharges. Environ. Pollut. 2006, 142, 295–302. [Google Scholar] [CrossRef]
  61. Bielen, A.; Šimatović, A.; Kosić-Vukšić, J.; Senta, I.; Ahel, M.; Babić, S.; Jurina, T.; González Plaza, J.J.; Milaković, M.; Udiković-Kolić, N. Negative Environmental Impacts of Antibiotic-Contaminated Effluents from Pharmaceutical Industries. Water Res. 2017, 126, 79–87. [Google Scholar] [CrossRef]
  62. Biel-Maeso, M.; Corada-Fernández, C.; Lara-Martín, P.A. Monitoring the Occurrence of Pharmaceuticals in Soils Irrigated with Reclaimed Wastewater. Environ. Pollut. 2018, 235, 312–321. [Google Scholar] [CrossRef]
  63. Blair, B.D.; Crago, J.P.; Hedman, C.J.; Treguer, R.J.F.; Magruder, C.; Royer, L.S.; Klaper, R.D. Evaluation of a Model for the Removal of Pharmaceuticals, Personal Care Products, and Hormones from Wastewater. Sci. Total Environ. 2013, 444, 515–521. [Google Scholar] [CrossRef] [PubMed]
  64. Bonvin, F.; Rutler, R.; Chèvre, N.; Halder, J.; Kohn, T. Spatial and Temporal Presence of a Wastewater-Derived Micropollutant Plume in Lake Geneva. Environ. Sci. Technol. 2011, 45, 4702–4709. [Google Scholar] [CrossRef] [PubMed]
  65. Martínez Bueno, M.J.; Agüera, A.; Gómez, M.J.; Hernando, M.D.; García-Reyes, J.F.; Fernández-Alba, A.R. Application of Liquid Chromatography/Quadrupole-Linear Ion Trap Mass Spectrometry and Time-of-Flight Mass Spectrometry to the Determination of Pharmaceuticals and Related Contaminants in Wastewater. Anal. Chem. 2007, 79, 9372–9384. [Google Scholar] [CrossRef]
  66. Butkovskyi, A.; Hernandez Leal, L.; Rijnaarts, H.H.M.; Zeeman, G. Fate of Pharmaceuticals in Full-Scale Source Separated Sanitation System. Water Res. 2015, 85, 384–392. [Google Scholar] [CrossRef]
  67. Calamari, D.; Zuccato, E.; Castiglioni, S.; Bagnati, R.; Fanelli, R. Strategic Survey of Therapeutic Drugs in the Rivers Po and Lambro in Northern Italy. Environ. Sci. Technol. 2003, 37, 1241–1248. [Google Scholar] [CrossRef]
  68. Carpenter, C.M.G.; Helbling, D.E. Widespread Micropollutant Monitoring in the Hudson River Estuary Reveals Spatiotemporal Micropollutant Clusters and Their Sources. Environ. Sci. Technol. 2018, 52, 6187–6196. [Google Scholar] [CrossRef]
  69. Castiglioni, S.; Bagnati, R.; Calamari, D.; Fanelli, R.; Zuccato, E. A Multiresidue Analytical Method Using Solid-Phase Extraction and High-Pressure Liquid Chromatography Tandem Mass Spectrometry to Measure Pharmaceuticals of Different Therapeutic Classes in Urban Wastewaters. J. Chromatogr. A 2005, 1092, 206–215. [Google Scholar] [CrossRef]
  70. Cheng, D.; Liu, X.; Zhao, S.; Cui, B.; Bai, J.; Li, Z. Influence of the Natural Colloids on the Multi-Phase Distributions of Antibiotics in the Surface Water from the Largest Lake in North China. Sci. Total Environ. 2017, 578, 649–659. [Google Scholar] [CrossRef]
  71. Christian, T.; Schneider, R.J.; Färber, H.A.; Skutlarek, D.; Meyer, M.T.; Goldbach, H.E. Determination of Antibiotic Residues in Manure, Soil, and Surface Waters. Acta Hydrochim. Hydrobiol. 2003, 31, 36–44. [Google Scholar] [CrossRef]
  72. Chung, H.S.; Choi, J.; Abd El-Aty, A.M.; Lee, Y.; Lee, H.S.; Kim, S.; Jung, H.; Kang, T.; Shin, H.; Shim, J. Simultaneous Determination of Seven Multiclass Veterinary Antibiotics in Surface Water Samples in the Republic of Korea Using Liquid Chromatography with Tandem Mass Spectrometry. J. Sep. Sci. 2016, 39, 4688–4699. [Google Scholar] [CrossRef]
  73. Ata, R.; Yıldız Töre, G. Characterization and Removal of Antibiotic Residues by NFC-Doped Photocatalytic Oxidation from Domestic and Industrial Secondary Treated Wastewaters in Meric-Ergene Basin and Reuse Assessment for Irrigation. J. Environ. Manag. 2019, 233, 673–680. [Google Scholar] [CrossRef]
  74. Čizmić, M.; Babić, S.; Kaštelan-Macan, M. Multi-Class Determination of Pharmaceuticals in Wastewaters by Solid-Phase Extraction and Liquid Chromatography Tandem Mass Spectrometry with Matrix Effect Study. Environ. Sci. Pollut. Res. 2017, 24, 20521–20539. [Google Scholar] [CrossRef]
  75. Coetsier, C.M.; Spinelli, S.; Lin, L.; Roig, B.; Touraud, E. Discharge of Pharmaceutical Products (PPs) through a Conventional Biological Sewage Treatment Plant: MECs vs PECs? Environ. Int. 2009, 35, 787–792. [Google Scholar] [CrossRef]
  76. Cui, J.; Zhang, K.; Huang, Q.; Yu, Y.; Peng, X. An Indirect Competitive Enzyme-Linked Immunosorbent Assay for Determination of Norfloxacin in Waters Using a Specific Polyclonal Antibody. Anal. Chim. Acta 2011, 688, 84–89. [Google Scholar] [CrossRef]
  77. Da Le, N.; Hoang, A.Q.; Hoang, T.T.H.; Nguyen, T.A.H.; Duong, T.T.; Pham, T.M.H.; Nguyen, T.D.; Hoang, V.C.; Phung, T.X.B.; Le, H.T.; et al. Antibiotic and Antiparasitic Residues in Surface Water of Urban Rivers in the Red River Delta (Hanoi, Vietnam): Concentrations, Profiles, Source Estimation, and Risk Assessment. Environ. Sci. Pollut. Res. 2021, 28, 10622–10632. [Google Scholar] [CrossRef]
  78. De Jesus Gaffney, V.; Cardoso, V.V.; Cardoso, E.; Teixeira, A.P.; Martins, J.; Benoliel, M.J.; Almeida, C.M.M. Occurrence and Behaviour of Pharmaceutical Compounds in a Portuguese Wastewater Treatment Plant: Removal Efficiency through Conventional Treatment Processes. Environ. Sci. Pollut. Res. 2017, 24, 14717–14734. [Google Scholar] [CrossRef]
  79. Tuc Dinh, Q.; Alliot, F.; Moreau-Guigon, E.; Eurin, J.; Chevreuil, M.; Labadie, P. Measurement of Trace Levels of Antibiotics in River Water Using On-Line Enrichment and Triple-Quadrupole LC–MS/MS. Talanta 2011, 85, 1238–1245. [Google Scholar] [CrossRef]
  80. Farrokh Shad, M.; Juby, G.J.G.; Delagah, S.; Sharbatmaleki, M. Evaluating Occurrence of Contaminants of Emerging Concerns in MF/RO Treatment of Primary Effluent for Water Reuse—Pilot Study. J. Water Reuse Desalination 2019, 9, 350–371. [Google Scholar] [CrossRef]
  81. Feitosa-Felizzola, J.; Chiron, S. Occurrence and Distribution of Selected Antibiotics in a Small Mediterranean Stream (Arc River, Southern France). J. Hydrol. 2009, 364, 50–57. [Google Scholar] [CrossRef]
  82. Fenech, C.; Nolan, K.; Rock, L.; Morrissey, A. An SPE LC–MS/MS Method for the Analysis of Human and Veterinary Chemical Markers within Surface Waters: An Environmental Forensics Application. Environ. Pollut. 2013, 181, 250–256. [Google Scholar] [CrossRef]
  83. Ferdig, M.; Kaleta, A.; Buchberger, W. Improved Liquid Chromatographic Determination of Nine Currently Used (Fluoro)Quinolones with Fluorescence and Mass Spectrometric Detection for Environmental Samples. J. Sep. Sci. 2005, 28, 1448–1456. [Google Scholar] [CrossRef]
  84. Fick, J.; Lindberg, R.H.; Kaj, L.; Brorström-Lundén, E. Results from the Swedish National Screening Programme 2010; IVL Swedish Environmental Research Institute: Stockholm, Sweden, 2011; p. 56. [Google Scholar]
  85. Finnegan, D.P.; Simonson, L.A.; Meyer, M.T. Occurrence of Antibiotic Compounds in Source Water and Finished Drinking Water from the Upper Scioto River Basin, Ohio, 2005–6; U.S. Geological Survey Scientific Investigations Report; U.S. Geological Survey: Reston, VA, USA, 2010; p. 16. [Google Scholar]
  86. Fonseca, E.; Hernández, F.; Ibáñez, M.; Rico, A.; Pitarch, E.; Bijlsma, L. Occurrence and Ecological Risks of Pharmaceuticals in a Mediterranean River in Eastern Spain. Environ. Int. 2020, 144, 106004. [Google Scholar] [CrossRef]
  87. Gbylik-Sikorska, M.; Posyniak, A.; Mitrowska, K.; Gajda, A.; Błądek, T.; Śniegocki, T.; Żmudzki, J. Occurrence of Veterinary Antibiotics and Chemotherapeutics in Fresh Water, Sediment, and Fish of the Rivers and Lakes in Poland. Bull. Vet. Inst. Pulawy 2014, 58, 399–404. [Google Scholar] [CrossRef]
  88. Ghosh, G.C.; Okuda, T.; Yamashita, N.; Tanaka, H. Occurrence and Elimination of Antibiotics at Four Sewage Treatment Plants in Japan and Their Effects on Bacterial Ammonia Oxidation. Water Sci. Technol. 2009, 59, 779–786. [Google Scholar] [CrossRef]
  89. Golet, E.M.; Alder, A.C.; Hartmann, A.; Ternes, T.A.; Giger, W. Trace Determination of Fluoroquinolone Antibacterial Agents in Urban Wastewater by Solid-Phase Extraction and Liquid Chromatography with Fluorescence Detection. Anal. Chem. 2001, 73, 3632–3638. [Google Scholar] [CrossRef]
  90. Golovko, O.; Kumar, V.; Fedorova, G.; Randak, T.; Grabic, R. Seasonal Changes in Antibiotics, Antidepressants/Psychiatric Drugs, Antihistamines and Lipid Regulators in a Wastewater Treatment Plant. Chemosphere 2014, 111, 418–426. [Google Scholar] [CrossRef]
  91. Grabic, R.; Fick, J.; Lindberg, R.H.; Fedorova, G.; Tysklind, M. Multi-Residue Method for Trace Level Determination of Pharmaceuticals in Environmental Samples Using Liquid Chromatography Coupled to Triple Quadrupole Mass Spectrometry. Talanta 2012, 100, 183–195. [Google Scholar] [CrossRef]
  92. Gracia-Lor, E.; Sancho, J.V.; Hernández, F. Multi-Class Determination of around 50 Pharmaceuticals, Including 26 Antibiotics, in Environmental and Wastewater Samples by Ultra-High Performance Liquid Chromatography–Tandem Mass Spectrometry. J. Chromatogr. A 2011, 1218, 2264–2275. [Google Scholar] [CrossRef]
  93. Gros, M.; Petrović, M.; Barceló, D. Tracing Pharmaceutical Residues of Different Therapeutic Classes in Environmental Waters by Using Liquid Chromatography/Quadrupole-Linear Ion Trap Mass Spectrometry and Automated Library Searching. Anal. Chem. 2009, 81, 898–912. [Google Scholar] [CrossRef]
  94. He, X.; Wang, Z.; Nie, X.; Yang, Y.; Pan, D.; Leung, A.O.W.; Cheng, Z.; Yang, Y.; Li, K.; Chen, K. Residues of Fluoroquinolones in Marine Aquaculture Environment of the Pearl River Delta, South China. Environ. Geochem. Health 2012, 34, 323–335. [Google Scholar] [CrossRef]
  95. Iglesias, A.; Nebot, C.; Vázquez, B.; Coronel-Olivares, C.; Abuín, C.; Cepeda, A. Monitoring the Presence of 13 Active Compounds in Surface Water Collected from Rural Areas in Northwestern Spain. Int. J. Environ. Res. Public. Health 2014, 11, 5251–5272. [Google Scholar] [CrossRef]
  96. Jiang, J.-Q.; Zhou, Z.; Sharma, V.K. Occurrence, Transportation, Monitoring and Treatment of Emerging Micro-Pollutants in Waste Water—A Review from Global Views. Microchem. J. 2013, 110, 292–300. [Google Scholar] [CrossRef]
  97. Fang, Y.; Karnjanapiboonwong, A.; Chase, D.A.; Wang, J.; Morse, A.N.; Anderson, T.A. Occurrence, Fate, and Persistence of Gemfibrozil in Water and Soil. Environ. Toxicol. Chem. 2012, 31, 550–555. [Google Scholar] [CrossRef]
  98. Kibuye, F.A.; Gall, H.E.; Elkin, K.R.; Ayers, B.; Veith, T.L.; Miller, M.; Jacob, S.; Hayden, K.R.; Watson, J.E.; Elliott, H.A. Fate of Pharmaceuticals in a Spray-Irrigation System: From Wastewater to Groundwater. Sci. Total Environ. 2019, 654, 197–208. [Google Scholar] [CrossRef]
  99. Kim, J.-W.; Jang, H.-S.; Kim, J.-G.; Ishibashi, H.; Hirano, M.; Nasu, K.; Ichikawa, N.; Takao, Y.; Shinohara, R.; Arizono, K. Occurrence of Pharmaceutical and Personal Care Products (PPCPs) in Surface Water from Mankyung River, South Korea. J. Health Sci. 2009, 55, 249–258. [Google Scholar] [CrossRef]
  100. Köck-Schulmeyer, M.; Ginebreda, A.; Postigo, C.; López-Serna, R.; Pérez, S.; Brix, R.; Llorca, M.; Alda, M.L.D.; Petrović, M.; Munné, A.; et al. Wastewater Reuse in Mediterranean Semi-Arid Areas: The Impact of Discharges of Tertiary Treated Sewage on the Load of Polar Micro Pollutants in the Llobregat River (NE Spain). Chemosphere 2011, 82, 670–678. [Google Scholar] [CrossRef]
  101. Kot-Wasik, A.; Jakimska, A.; Śliwka-Kaszyńska, M. Occurrence and Seasonal Variations of 25 Pharmaceutical Residues in Wastewater and Drinking Water Treatment Plants. Environ. Monit. Assess. 2016, 188, 661. [Google Scholar] [CrossRef]
  102. Krakkó, D.; Licul-Kucera, V.; Záray, G.; Mihucz, V.G. Single-Run Ultra-High Performance Liquid Chromatography for Quantitative Determination of Ultra-Traces of Ten Popular Active Pharmaceutical Ingredients by Quadrupole Time-of-Flight Mass Spectrometry after Offline Preconcentration by Solid Phase Extraction from Drinking and River Waters as Well as Treated Wastewater. Microchem. J. 2019, 148, 108–119. [Google Scholar] [CrossRef]
  103. Lara-Martín, P.A.; González-Mazo, E.; Petrovic, M.; Barceló, D.; Brownawell, B.J. Occurrence, Distribution and Partitioning of Nonionic Surfactants and Pharmaceuticals in the Urbanized Long Island Sound Estuary (NY). Mar. Pollut. Bull. 2014, 85, 710–719. [Google Scholar] [CrossRef]
  104. Lépesová, K.; Kraková, L.; Pangallo, D.; Medveďová, A.; Olejníková, P.; Mackuľak, T.; Tichý, J.; Grabic, R.; Birošová, L. Prevalence of Antibiotic-Resistant Coliform Bacteria, Enterococcus Spp. and Staphylococcus Spp. in Wastewater Sewerage Biofilm. J. Glob. Antimicrob. Resist. 2018, 14, 145–151. [Google Scholar] [CrossRef]
  105. Li, W.; Shi, Y.; Gao, L.; Liu, J.; Cai, Y. Occurrence of Antibiotics in Water, Sediments, Aquatic Plants, and Animals from Baiyangdian Lake in North China. Chemosphere 2012, 89, 1307–1315. [Google Scholar] [CrossRef]
  106. Lin, A.Y.-C.; Yu, T.-H.; Lin, C.-F. Pharmaceutical Contamination in Residential, Industrial, and Agricultural Waste Streams: Risk to Aqueous Environments in Taiwan. Chemosphere 2008, 74, 131–141. [Google Scholar] [CrossRef]
  107. Lin, H.; Li, H.; Chen, L.; Li, L.; Yin, L.; Lee, H.; Yang, Z. Mass Loading and Emission of Thirty-Seven Pharmaceuticals in a Typical Municipal Wastewater Treatment Plant in Hunan Province, Southern China. Ecotoxicol. Environ. Saf. 2018, 147, 530–536. [Google Scholar] [CrossRef]
  108. Lindberg, R.H.; Olofsson, U.; Rendahl, P.; Johansson, M.I.; Tysklind, M.; Andersson, B.A.V. Behavior of Fluoroquinolones and Trimethoprim during Mechanical, Chemical, and Active Sludge Treatment of Sewage Water and Digestion of Sludge. Environ. Sci. Technol. 2006, 40, 1042–1048. [Google Scholar] [CrossRef]
  109. European Commission; Joint Research Centre; Institute for Environment and Sustainability. EU Wide Monitoring Survey on Waste Water Treatment Plant Effluents.; Publications Office: Luxembourg, 2012. [Google Scholar]
  110. Loper, C.A.; Crawford, J.K.; Otto, K.L.; Manning, R.L.; Meyer, M.T.; Furlong, E.T. Concentrations of Selected Pharmaceuticals and Antibiotics in South-Central Pennsylvania Waters, March through September 2006; USGS Publications Warehouse: Reston, VA, USA, 2007. [Google Scholar]
  111. López-Serna, R.; Petrović, M.; Barceló, D. Development of a Fast Instrumental Method for the Analysis of Pharmaceuticals in Environmental and Wastewaters Based on Ultra High Performance Liquid Chromatography (UHPLC)–Tandem Mass Spectrometry (MS/MS). Chemosphere 2011, 85, 1390–1399. [Google Scholar] [CrossRef]
  112. Luo, Y.; Xu, L.; Rysz, M.; Wang, Y.; Zhang, H.; Alvarez, P.J.J. Occurrence and Transport of Tetracycline, Sulfonamide, Quinolone, and Macrolide Antibiotics in the Haihe River Basin, China. Environ. Sci. Technol. 2011, 45, 1827–1833. [Google Scholar] [CrossRef]
  113. Ma, N.; Tong, L.; Li, Y.; Yang, C.; Tan, Q.; He, J. Distribution of Antibiotics in Lake Water-Groundwater—Sediment System in Chenhu Lake Area. Environ. Res. 2022, 204, 112343. [Google Scholar] [CrossRef]
  114. Bueno, M.J.M.; Gomez, M.J.; Herrera, S.; Hernando, M.D.; Agüera, A.; Fernández-Alba, A.R. Occurrence and Persistence of Organic Emerging Contaminants and Priority Pollutants in Five Sewage Treatment Plants of Spain: Two Years Pilot Survey Monitoring. Environ. Pollut. 2012, 164, 267–273. [Google Scholar] [CrossRef]
  115. Massey, L.B.; Haggard, B.E.; Galloway, J.M.; Loftin, K.A.; Meyer, M.T.; Green, W.R. Antibiotic Fate and Transport in Three Effluent-Dominated Ozark Streams. Ecol. Eng. 2010, 36, 930–938. [Google Scholar] [CrossRef]
  116. Minh, T.B.; Leung, H.W.; Loi, I.H.; Chan, W.H.; So, M.K.; Mao, J.Q.; Choi, D.; Lam, J.C.W.; Zheng, G.; Martin, M.; et al. Antibiotics in the Hong Kong Metropolitan Area: Ubiquitous Distribution and Fate in Victoria Harbour. Mar. Pollut. Bull. 2009, 58, 1052–1062. [Google Scholar] [CrossRef]
  117. Miossec, C.; Lanceleur, L.; Monperrus, M. Multi-residue Analysis of 44 Pharmaceutical Compounds in Environmental Water Samples by Solid-phase Extraction Coupled to Liquid Chromatography-tandem Mass Spectrometry. J. Sep. Sci. 2019, 42, 1853–1866. [Google Scholar] [CrossRef]
  118. Mompelat, S.; Thomas, O.; Le Bot, B. Contamination Levels of Human Pharmaceutical Compounds in French Surface and Drinking Water. J. Environ. Monit. 2011, 13, 2929. [Google Scholar] [CrossRef]
  119. Morosini, C.; Marsoni, M.; Torretta, V.; Conti, F.; Ragazzi, M.; Rada, E.; Cioca, G. Factors Affecting Spatial and Temporal Concentration Variability of Pharmaceuticals: Comparison between Two WWTPs. Sustainability 2017, 9, 1466. [Google Scholar] [CrossRef]
  120. Mu, R.; Shi, H.; Adams, C.; Eichholz, T.; Ma, Y. Detection, Occurrence, and Removal of Selected Pharmaceuticals in Missouri Source and Finished Drinking Waters. Urban Water J. 2017, 14, 704–712. [Google Scholar] [CrossRef]
  121. Mutiyar, P.K.; Mittal, A.K. Risk Assessment of Antibiotic Residues in Different Water Matrices in India: Key Issues and Challenges. Environ. Sci. Pollut. Res. 2014, 21, 7723–7736. [Google Scholar] [CrossRef] [PubMed]
  122. Nageswara Rao, R.; Venkateswarlu, N.; Narsimha, R. Determination of Antibiotics in Aquatic Environment by Solid-Phase Extraction Followed by Liquid Chromatography–Electrospray Ionization Mass Spectrometry. J. Chromatogr. A 2008, 1187, 151–164. [Google Scholar] [CrossRef] [PubMed]
  123. Nakada, N.; Komori, K.; Suzuki, Y.; Konishi, C.; Houwa, I.; Tanaka, H. Occurrence of 70 Pharmaceutical and Personal Care Products in Tone River Basin in Japan. Water Sci. Technol. 2007, 56, 133–140. [Google Scholar] [CrossRef] [PubMed]
  124. Panthi, S.; Sapkota, A.R.; Raspanti, G.; Allard, S.M.; Bui, A.; Craddock, H.A.; Murray, R.; Zhu, L.; East, C.; Handy, E.; et al. Pharmaceuticals, Herbicides, and Disinfectants in Agricultural Water Sources. Environ. Res. 2019, 174, 1–8. [Google Scholar] [CrossRef] [PubMed]
  125. Papageorgiou, M.; Kosma, C.; Lambropoulou, D. Seasonal Occurrence, Removal, Mass Loading and Environmental Risk Assessment of 55 Pharmaceuticals and Personal Care Products in a Municipal Wastewater Treatment Plant in Central Greece. Sci. Total Environ. 2016, 543, 547–569. [Google Scholar] [CrossRef] [PubMed]
  126. Pasquini, L.; Munoz, J.-F.; Pons, M.-N.; Yvon, J.; Dauchy, X.; France, X.; Le, N.D.; France-Lanord, C.; Görner, T. Occurrence of Eight Household Micropollutants in Urban Wastewater and Their Fate in a Wastewater Treatment Plant. Statistical Evaluation. Sci. Total Environ. 2014, 481, 459–468. [Google Scholar] [CrossRef] [PubMed]
  127. Wahlberg, C.; Björlenius, B.; Paxéus, N. Läkemedelsrester i Stockholms vattenmiljö; Stockholm Vatten: Stockholm, Sweden, 2010; p. 46. [Google Scholar]
  128. Pena, A.; Chmielova, D.; Lino, C.M.; Solich, P. Determination of Fluoroquinolone Antibiotics in Surface Waters from Mondego River by High Performance Liquid Chromatography Using a Monolithic Column. J. Sep. Sci. 2007, 30, 2924–2928. [Google Scholar] [CrossRef] [PubMed]
  129. Peng, X.; Zhang, K.; Tang, C.; Huang, Q.; Yu, Y.; Cui, J. Distribution Pattern, Behavior, and Fate of Antibacterials in Urban Aquatic Environments in South China. J. Environ. Monit. 2011, 13, 446–454. [Google Scholar] [CrossRef]
  130. Pereira, A.M.P.T.; Silva, L.J.G.; Lino, C.M.; Meisel, L.M.; Pena, A. Assessing Environmental Risk of Pharmaceuticals in Portugal: An Approach for the Selection of the Portuguese Monitoring Stations in Line with Directive 2013/39/EU. Chemosphere 2016, 144, 2507–2515. [Google Scholar] [CrossRef] [PubMed]
  131. Pinnekamp, J.; Beier, S.; Cramer, C.; Schröderh, H.; Mauer, C.; Selke, D. Eliminierung von Spurenstoffen Aus Krankenhausabwässern Mit Membrantechnik Und Weitergehenden Behandlungsverfahren—Pilotprojekt Kreiskrankenhaus Waldbröl; RWTH Aachen: Aachen, Germany, 2009; p. 232. [Google Scholar]
  132. Prieto, A.; Schrader, S.; Bauer, C.; Möder, M. Synthesis of a Molecularly Imprinted Polymer and Its Application for Microextraction by Packed Sorbent for the Determination of Fluoroquinolone Related Compounds in Water. Anal. Chim. Acta 2011, 685, 146–152. [Google Scholar] [CrossRef] [PubMed]
  133. Rico, A.; Oliveira, R.; McDonough, S.; Matser, A.; Khatikarn, J.; Satapornvanit, K.; Nogueira, A.J.A.; Soares, A.M.V.M.; Domingues, I.; Van Den Brink, P.J. Use, Fate and Ecological Risks of Antibiotics Applied in Tilapia Cage Farming in Thailand. Environ. Pollut. 2014, 191, 8–16. [Google Scholar] [CrossRef] [PubMed]
  134. Rico, A.; Arenas-Sánchez, A.; Alonso-Alonso, C.; López-Heras, I.; Nozal, L.; Rivas-Tabares, D.; Vighi, M. Identification of Contaminants of Concern in the Upper Tagus River Basin (Central Spain). Part 1: Screening, Quantitative Analysis and Comparison of Sampling Methods. Sci. Total Environ. 2019, 666, 1058–1070. [Google Scholar] [CrossRef] [PubMed]
  135. Rodriguez-Mozaz, S.; Vaz-Moreira, I.; Varela Della Giustina, S.; Llorca, M.; Barceló, D.; Schubert, S.; Berendonk, T.U.; Michael-Kordatou, I.; Fatta-Kassinos, D.; Martinez, J.L.; et al. Antibiotic Residues in Final Effluents of European Wastewater Treatment Plants and Their Impact on the Aquatic Environment. Environ. Int. 2020, 140, 105733. [Google Scholar] [CrossRef] [PubMed]
  136. Rosal, R.; Rodríguez, A.; Perdigón-Melón, J.A.; Petre, A.; García-Calvo, E.; Gómez, M.J.; Agüera, A.; Fernández-Alba, A.R. Occurrence of Emerging Pollutants in Urban Wastewater and Their Removal through Biological Treatment Followed by Ozonation. Water Res. 2010, 44, 578–588. [Google Scholar] [CrossRef] [PubMed]
  137. Santos, L.H.M.L.M.; Gros, M.; Rodriguez-Mozaz, S.; Delerue-Matos, C.; Pena, A.; Barceló, D.; Montenegro, M.C.B.S.M. Contribution of Hospital Effluents to the Load of Pharmaceuticals in Urban Wastewaters: Identification of Ecologically Relevant Pharmaceuticals. Sci. Total Environ. 2013, 461–462, 302–316. [Google Scholar] [CrossRef] [PubMed]
  138. Sharma, B.M.; Bečanová, J.; Scheringer, M.; Sharma, A.; Bharat, G.K.; Whitehead, P.G.; Klánová, J.; Nizzetto, L. Health and Ecological Risk Assessment of Emerging Contaminants (Pharmaceuticals, Personal Care Products, and Artificial Sweeteners) in Surface and Groundwater (Drinking Water) in the Ganges River Basin, India. Sci. Total Environ. 2019, 646, 1459–1467. [Google Scholar] [CrossRef] [PubMed]
  139. Shi, Y.; Liu, J.; Zhuo, L.; Yan, X.; Cai, F.; Luo, W.; Ren, M.; Liu, Q.; Yu, Y. Antibiotics in Wastewater from Multiple Sources and Surface Water of the Yangtze River in Chongqing in China. Environ. Monit. Assess. 2020, 192, 159. [Google Scholar] [CrossRef]
  140. Sim, W.-J.; Lee, J.-W.; Oh, J.-E. Occurrence and Fate of Pharmaceuticals in Wastewater Treatment Plants and Rivers in Korea. Environ. Pollut. 2010, 158, 1938–1947. [Google Scholar] [CrossRef]
  141. Sousa, M.A.; Gonçalves, C.; Vilar, V.J.P.; Boaventura, R.A.R.; Alpendurada, M.F. Suspended TiO2-Assisted Photocatalytic Degradation of Emerging Contaminants in a Municipal WWTP Effluent Using a Solar Pilot Plant with CPCs. Chem. Eng. J. 2012, 198–199, 301–309. [Google Scholar] [CrossRef]
  142. Spongberg, A.L.; Witter, J.D. Pharmaceutical Compounds in the Wastewater Process Stream in Northwest Ohio. Sci. Total Environ. 2008, 397, 148–157. [Google Scholar] [CrossRef] [PubMed]
  143. Sturini, M.; Speltini, A.; Pretali, L.; Fasani, E.; Profumo, A. Solid-phase Extraction and HPLC Determination of Fluoroquinolones in Surface Waters. J. Sep. Sci. 2009, 32, 3020–3028. [Google Scholar] [CrossRef] [PubMed]
  144. Takasu, H.; Suzuki, S.; Reungsang, A.; Viet, P.H. Fluoroquinolone (FQ) Contamination Does Not Correlate with Occurrence of FQ-Resistant Bacteria in Aquatic Environments of Vietnam and Thailand. Microbes Environ. 2011, 26, 135–143. [Google Scholar] [CrossRef] [PubMed]
  145. Tamtam, F.; Mercier, F.; Le Bot, B.; Eurin, J.; Tuc Dinh, Q.; Clément, M.; Chevreuil, M. Occurrence and Fate of Antibiotics in the Seine River in Various Hydrological Conditions. Sci. Total Environ. 2008, 393, 84–95. [Google Scholar] [CrossRef] [PubMed]
  146. Tewari, S.; Jindal, R.; Kho, Y.L.; Eo, S.; Choi, K. Major Pharmaceutical Residues in Wastewater Treatment Plants and Receiving Waters in Bangkok, Thailand, and Associated Ecological Risks. Chemosphere 2013, 91, 697–704. [Google Scholar] [CrossRef] [PubMed]
  147. Tong, C.; Zhuo, X.; Guo, Y. Occurrence and Risk Assessment of Four Typical Fluoroquinolone Antibiotics in Raw and Treated Sewage and in Receiving Waters in Hangzhou, China. J. Agric. Food Chem. 2011, 59, 7303–7309. [Google Scholar] [CrossRef] [PubMed]
  148. Vazquez-Roig, P.; Andreu, V.; Onghena, M.; Blasco, C.; Picó, Y. Assessment of the Occurrence and Distribution of Pharmaceuticals in a Mediterranean Wetland (L’Albufera, Valencia, Spain) by LC-MS/MS. Anal. Bioanal. Chem. 2011, 400, 1287–1301. [Google Scholar] [CrossRef] [PubMed]
  149. Vazquez-Roig, P.; Andreu, V.; Blasco, C.; Picó, Y. Risk Assessment on the Presence of Pharmaceuticals in Sediments, Soils and Waters of the Pego–Oliva Marshlands (Valencia, Eastern Spain). Sci. Total Environ. 2012, 440, 24–32. [Google Scholar] [CrossRef]
  150. Velpandian, T.; Halder, N.; Nath, M.; Das, U.; Moksha, L.; Gowtham, L.; Batta, S.P. Un-Segregated Waste Disposal: An Alarming Threat of Antimicrobials in Surface and Ground Water Sources in Delhi. Environ. Sci. Pollut. Res. 2018, 25, 29518–29528. [Google Scholar] [CrossRef]
  151. Vergeynst, L.; Haeck, A.; De Wispelaere, P.; Van Langenhove, H.; Demeestere, K. Multi-Residue Analysis of Pharmaceuticals in Wastewater by Liquid Chromatography–Magnetic Sector Mass Spectrometry: Method Quality Assessment and Application in a Belgian Case Study. Chemosphere 2015, 119, S2–S8. [Google Scholar] [CrossRef] [PubMed]
  152. Vieno, N.M.; Tuhkanen, T.; Kronberg, L. Analysis of Neutral and Basic Pharmaceuticals in Sewage Treatment Plants and in Recipient Rivers Using Solid Phase Extraction and Liquid Chromatography–Tandem Mass Spectrometry Detection. J. Chromatogr. A 2006, 1134, 101–111. [Google Scholar] [CrossRef] [PubMed]
  153. Vulliet, E.; Cren-Olivé, C. Screening of Pharmaceuticals and Hormones at the Regional Scale, in Surface and Groundwaters Intended to Human Consumption. Environ. Pollut. 2011, 159, 2929–2934. [Google Scholar] [CrossRef]
  154. Wiest, L.; Chonova, T.; Bergé, A.; Baudot, R.; Bessueille-Barbier, F.; Ayouni-Derouiche, L.; Vulliet, E. Two-Year Survey of Specific Hospital Wastewater Treatment and Its Impact on Pharmaceutical Discharges. Environ. Sci. Pollut. Res. 2018, 25, 9207–9218. [Google Scholar] [CrossRef]
  155. Wu, M.; Que, C.; Tang, L.; Xu, H.; Xiang, J.; Wang, J.; Shi, W.; Xu, G. Distribution, Fate, and Risk Assessment of Antibiotics in Five Wastewater Treatment Plants in Shanghai, China. Environ. Sci. Pollut. Res. 2016, 23, 18055–18063. [Google Scholar] [CrossRef]
  156. Xiao, Y.; Chang, H.; Jia, A.; Hu, J. Trace Analysis of Quinolone and Fluoroquinolone Antibiotics from Wastewaters by Liquid Chromatography–Electrospray Tandem Mass Spectrometry. J. Chromatogr. A 2008, 1214, 100–108. [Google Scholar] [CrossRef]
  157. Xu, W.; Zhang, G.; Zou, S.; Li, X.; Liu, Y. Determination of Selected Antibiotics in the Victoria Harbour and the Pearl River, South China Using High-Performance Liquid Chromatography-Electrospray Ionization Tandem Mass Spectrometry. Environ. Pollut. 2007, 145, 672–679. [Google Scholar] [CrossRef]
  158. Yang, X.; Flowers, R.C.; Weinberg, H.S.; Singer, P.C. Occurrence and Removal of Pharmaceuticals and Personal Care Products (PPCPs) in an Advanced Wastewater Reclamation Plant. Water Res. 2011, 45, 5218–5228. [Google Scholar] [CrossRef]
  159. Yao, L.; Wang, Y.; Tong, L.; Deng, Y.; Li, Y.; Gan, Y.; Guo, W.; Dong, C.; Duan, Y.; Zhao, K. Occurrence and Risk Assessment of Antibiotics in Surface Water and Groundwater from Different Depths of Aquifers: A Case Study at Jianghan Plain, Central China. Ecotoxicol. Environ. Saf. 2017, 135, 236–242. [Google Scholar] [CrossRef]
  160. Yu, Y.; Wu, G.; Wang, C.; Lu, N.; Yuan, X.; Zhu, X. Pollution Characteristics of Antibiotics and Antibiotic Resistance of Coliform Bacteria in the Yitong River, China. Environ. Monit. Assess. 2019, 191, 516. [Google Scholar] [CrossRef] [PubMed]
  161. Zhang, R.; Zhang, G.; Zheng, Q.; Tang, J.; Chen, Y.; Xu, W.; Zou, Y.; Chen, X. Occurrence and Risks of Antibiotics in the Laizhou Bay, China: Impacts of River Discharge. Ecotoxicol. Environ. Saf. 2012, 80, 208–215. [Google Scholar] [CrossRef]
  162. Zhang, H.; Liu, P.; Feng, Y.; Yang, F. Fate of Antibiotics during Wastewater Treatment and Antibiotic Distribution in the Effluent-Receiving Waters of the Yellow Sea, Northern China. Mar. Pollut. Bull. 2013, 73, 282–290. [Google Scholar] [CrossRef]
  163. Zhou, J.L.; Maskaoui, K.; Lufadeju, A. Optimization of Antibiotic Analysis in Water by Solid-Phase Extraction and High Performance Liquid Chromatography–Mass Spectrometry/Mass Spectrometry. Anal. Chim. Acta 2012, 731, 32–39. [Google Scholar] [CrossRef]
  164. Zou, S.; Xu, W.; Zhang, R.; Tang, J.; Chen, Y.; Zhang, G. Occurrence and Distribution of Antibiotics in Coastal Water of the Bohai Bay, China: Impacts of River Discharge and Aquaculture Activities. Environ. Pollut. 2011, 159, 2913–2920. [Google Scholar] [CrossRef]
  165. Zuccato, E.; Castiglioni, S.; Fanelli, R. Identification of the Pharmaceuticals for Human Use Contaminating the Italian Aquatic Environment. J. Hazard. Mater. 2005, 122, 205–209. [Google Scholar] [CrossRef] [PubMed]
  166. 166. Nordic Council of Ministers. PPCP Monitoring in the Nordic Countries—Status Report. Nordic Council of Ministers: Copenhagen, Denmark, 2012.
  167. Barber, L.B.; Keefe, S.H.; Kolpin, D.W.; Schnoebelen, D.J.; Flynn, J.L.; Brown, G.K.; Furlong, E.T.; Glassmeyer, S.T.; Gray, J.L.; Meyer, M.T.; et al. Lagrangian Sampling of Wastewater Treatment Plant Effluent in Boulder Creek, Colorado, and Fourmile Creek, Iowa, during the Summer of 2003 and Spring of 2005—Hydrological and Water-Quality Data; Open-File Report; U.S. Geological Survey: Reston, VA, USA, 2011. [Google Scholar]
  168. Xu, Y.; Chen, T.; Wang, Y.; Tao, H.; Liu, S.; Shi, W. The Occurrence and Removal of Selected Fluoroquinolones in Urban Drinking Water Treatment Plants. Environ. Monit. Assess. 2015, 187, 729. [Google Scholar] [CrossRef] [PubMed]
  169. Benito-Peña, E.; Urraca, J.L.; Sellergren, B.; Moreno-Bondi, M.C. Solid-Phase Extraction of Fluoroquinolones from Aqueous Samples Using a Water-Compatible Stochiometrically Imprinted Polymer. J. Chromatogr. A 2008, 1208, 62–70. [Google Scholar] [CrossRef]
  170. Chen, L.; Zhang, X.; Xu, Y.; Du, X.; Sun, X.; Sun, L.; Wang, H.; Zhao, Q.; Yu, A.; Zhang, H.; et al. Determination of Fluoroquinolone Antibiotics in Environmental Water Samples Based on Magnetic Molecularly Imprinted Polymer Extraction Followed by Liquid Chromatography–Tandem Mass Spectrometry. Anal. Chim. Acta 2010, 662, 31–38. [Google Scholar] [CrossRef]
  171. Azevedo, F.C.R.; Vaz, I.C.D.; Barbosa, F.A.R.; Magalhães, S.M.S. Toxicological Effects of Ciprofloxacin and Chlorhexidine on Growth and Chlorophylla Synthesis of Freshwater Cyanobacteria. Braz. J. Pharm. Sci. 2019, 55, e17661. [Google Scholar] [CrossRef]
  172. Angeles, L.F.; Islam, S.; Aldstadt, J.; Saqeeb, K.N.; Alam, M.; Khan, M.A.; Johura, F.-T.; Ahmed, S.I.; Aga, D.S. Retrospective Suspect Screening Reveals Previously Ignored Antibiotics, Antifungal Compounds, and Metabolites in Bangladesh Surface Waters. Sci. Total Environ. 2020, 712, 136285. [Google Scholar] [CrossRef]
  173. Su, D.; Ben, W.; Strobel, B.W.; Qiang, Z. Impacts of Wastewater Treatment Plant Upgrades on the Distribution and Risks of Pharmaceuticals in Receiving Rivers. J. Hazard. Mater. 2021, 406, 124331. [Google Scholar] [CrossRef] [PubMed]
  174. Ding, G.; Chen, G.; Liu, Y.; Li, M.; Liu, X. Occurrence and Risk Assessment of Fluoroquinolone Antibiotics in Reclaimed Water and Receiving Groundwater with Different Replenishment Pathways. Sci. Total Environ. 2020, 738, 139802. [Google Scholar] [CrossRef] [PubMed]
  175. Zhang, L.; Bai, J.; Zhang, K.; Wang, Y.; Xiao, R.; Campos, M.; Acuña, J.; Jorquera, M.A. Occurrence, Bioaccumulation and Ecological Risks of Antibiotics in the Water-Plant-Sediment Systems in Different Functional Areas of the Largest Shallow Lake in North China: Impacts of River Input and Historical Agricultural Activities. Sci. Total Environ. 2023, 857, 159260. [Google Scholar] [CrossRef] [PubMed]
  176. Lu, S.; Lin, C.; Lei, K.; Xin, M.; Gu, X.; Lian, M.; Wang, B.; Liu, X.; Ouyang, W.; He, M. Profiling of the Spatiotemporal Distribution, Risks, and Prioritization of Antibiotics in the Waters of Laizhou Bay, Northern China. J. Hazard. Mater. 2022, 424, 127487. [Google Scholar] [CrossRef] [PubMed]
  177. Zhou, Q.; Liu, G.; Arif, M.; Shi, X.; Wang, S. Occurrence and Risk Assessment of Antibiotics in the Surface Water of Chaohu Lake and Its Tributaries in China. Sci. Total Environ. 2022, 807, 151040. [Google Scholar] [CrossRef]
  178. Jiang, Y.; Li, M.; Guo, C.; An, D.; Xu, J.; Zhang, Y.; Xi, B. Distribution and Ecological Risk of Antibiotics in a Typical Effluent–Receiving River (Wangyang River) in North China. Chemosphere 2014, 112, 267–274. [Google Scholar] [CrossRef]
  179. Guo, X.; Song, R.; Lu, S.; Liu, X.; Chen, J.; Wan, Z.; Bi, B. Multi-Media Occurrence of Antibiotics and Antibiotic Resistance Genes in East Dongting Lake. Front. Environ. Sci. 2022, 10, 866332. [Google Scholar] [CrossRef]
  180. Chen, Y.; Xie, Q.; Wan, J.; Yang, S.; Wang, Y.; Fan, H. Occurrence and Risk Assessment of Antibiotics in Multifunctional Reservoirs in Dongguan, China. Environ. Sci. Pollut. Res. 2020, 27, 13565–13574. [Google Scholar] [CrossRef] [PubMed]
  181. Chen, J.; Huang, L.; Wang, Q.; Zeng, H.; Xu, J.; Chen, Z. Antibiotics in Aquaculture Ponds from Guilin, South of China: Occurrence, Distribution, and Health Risk Assessment. Environ. Res. 2022, 204, 112084. [Google Scholar] [CrossRef]
  182. Wang, C.; Lu, Y.; Sun, B.; Zhang, M.; Wang, R.; Li, X.; Mao, R.; Cao, Z.; Song, S. Contamination, Transport, and Ecological Risks of Pharmaceuticals and Personal Care Products in a Large Irrigation Region. Sci. Total Environ. 2022, 851, 158179. [Google Scholar] [CrossRef]
  183. Liu, J.; Deng, W.-J.; Ying, G.-G.; Tsang, E.P.K.; Hong, H.-C. Occurrence and Distribution of Antibiotics in Surface Water. Ecotoxicology 2022, 31, 1111–1119. [Google Scholar] [CrossRef] [PubMed]
  184. Zhang, G.; Liu, X.; Lu, S.; Zhang, J.; Wang, W. Occurrence of Typical Antibiotics in Nansi Lake’s Inflowing Rivers and Antibiotic Source Contribution to Nansi Lake Based on Principal Component Analysis-Multiple Linear Regression Model. Chemosphere 2020, 242, 125269. [Google Scholar] [CrossRef] [PubMed]
  185. Wang, C.; Zhao, Y.; Liu, S.; Xiao, Q.; Liang, W.; Song, Y. Contamination, Distribution, and Risk Assessment of Antibiotics in the Urban Surface Water of the Pearl River in Guangzhou, South China. Environ. Monit. Assess. 2021, 193, 98. [Google Scholar] [CrossRef] [PubMed]
  186. Yang, J.; Huang, Y.; Chen, Y.; Hassan, M.; Zhang, X.; Zhang, B.; Gin, K.Y.-H.; He, Y. Multi-Phase Distribution, Spatiotemporal Variation and Risk Assessment of Antibiotics in a Typical Urban-Rural Watershed. Ecotoxicol. Environ. Saf. 2020, 206, 111156. [Google Scholar] [CrossRef] [PubMed]
  187. Wu, Y.; Song, S.; Chen, X.; Shi, Y.; Cui, H.; Liu, Y.; Yang, S. Source-Specific Ecological Risks and Critical Source Identification of PPCPs in Surface Water: Comparing Urban and Rural Areas. Sci. Total Environ. 2023, 854, 158792. [Google Scholar] [CrossRef] [PubMed]
  188. Ci, M.; Zhang, G.; Yan, X.; Dong, W.; Xu, W.; Wang, W.; Fan, Y. Occurrence of Antibiotics in the Xiaoqing River Basin and Antibiotic Source Contribution-a Case Study of Jinan City, China. Environ. Sci. Pollut. Res. 2021, 28, 25241–25254. [Google Scholar] [CrossRef] [PubMed]
  189. Li, J.; Cui, M.; Zhang, H. Spatial and Temporal Variations of Antibiotics in a Tidal River. Environ. Monit. Assess. 2020, 192, 336. [Google Scholar] [CrossRef] [PubMed]
  190. Wang, K.; Su, Z.; Reguyal, F.; Bian, R.; Li, W.; Yu, H.; Sun, Y.; Zhuang, Y.; Shang, W. Seasonal Occurrence of Multiple Classes of Antibiotics in East China Rivers and Their Association with Suspended Particulate Matter. Sci. Total Environ. 2022, 853, 158579. [Google Scholar] [CrossRef] [PubMed]
  191. Zhang, G.; Lu, S.; Wang, Y.; Liu, X.; Liu, Y.; Xu, J.; Zhang, T.; Wang, Z.; Yang, Y. Occurrence of Antibiotics and Antibiotic Resistance Genes and Their Correlations in Lower Yangtze River, China. Environ. Pollut. 2020, 257, 113365. [Google Scholar] [CrossRef]
  192. Feng, J.; Liu, Q.; Ru, X.; Xi, N.; Sun, J. Occurrence and Distribution of Priority Pharmaceuticals in the Yellow River and the Huai River in Henan, China. Environ. Sci. Pollut. Res. 2020, 27, 16816–16826. [Google Scholar] [CrossRef]
  193. Malev, O.; Babić, S.; Sima Cota, A.; Stipaničev, D.; Repec, S.; Drnić, M.; Lovrić, M.; Bojanić, K.; Radić Brkanac, S.; Čož-Rakovac, R.; et al. Combining Short-Term Bioassays Using Fish and Crustacean Model Organisms with ToxCast in Vitro Data and Broad-Spectrum Chemical Analysis for Environmental Risk Assessment of the River Water (Sava, Croatia). Environ. Pollut. 2022, 292, 118440. [Google Scholar] [CrossRef] [PubMed]
  194. Diwan, V.; Hanna, N.; Purohit, M.; Chandran, S.; Riggi, E.; Parashar, V.; Tamhankar, A.; Stålsby Lundborg, C. Seasonal Variations in Water-Quality, Antibiotic Residues, Resistant Bacteria and Antibiotic Resistance Genes of Escherichia coli Isolates from Water and Sediments of the Kshipra River in Central India. Int. J. Environ. Res. Public. Health 2018, 15, 1281. [Google Scholar] [CrossRef] [PubMed]
  195. Riva, F.; Zuccato, E.; Davoli, E.; Fattore, E.; Castiglioni, S. Risk Assessment of a Mixture of Emerging Contaminants in Surface Water in a Highly Urbanized Area in Italy. J. Hazard. Mater. 2019, 361, 103–110. [Google Scholar] [CrossRef] [PubMed]
  196. Zuccato, E.; Castiglioni, S.; Bagnati, R.; Melis, M.; Fanelli, R. Source, Occurrence and Fate of Antibiotics in the Italian Aquatic Environment. J. Hazard. Mater. 2010, 179, 1042–1048. [Google Scholar] [CrossRef] [PubMed]
  197. Adachi, F.; Yamamoto, A.; Takakura, K.-I.; Kawahara, R. Occurrence of Fluoroquinolones and Fluoroquinolone-Resistance Genes in the Aquatic Environment. Sci. Total Environ. 2013, 444, 508–514. [Google Scholar] [CrossRef] [PubMed]
  198. Fernandes, M.J.; Paíga, P.; Silva, A.; Llaguno, C.P.; Carvalho, M.; Vázquez, F.M.; Delerue-Matos, C. Antibiotics and Antidepressants Occurrence in Surface Waters and Sediments Collected in the North of Portugal. Chemosphere 2020, 239, 124729. [Google Scholar] [CrossRef] [PubMed]
  199. Arun, S.; Xin, L.; Gaonkar, O.; Neppolian, B.; Zhang, G.; Chakraborty, P. Antibiotics in Sewage Treatment Plants, Receiving Water Bodies and Groundwater of Chennai City and the Suburb, South India: Occurrence, Removal Efficiencies, and Risk Assessment. Sci. Total Environ. 2022, 851, 158195. [Google Scholar] [CrossRef] [PubMed]
  200. Singh, V.; Suthar, S. Occurrence, Seasonal Variations, and Ecological Risk of Pharmaceuticals and Personal Care Products in River Ganges at Two Holy Cities of India. Chemosphere 2021, 268, 129331. [Google Scholar] [CrossRef]
  201. Low, K.; Chai, L.; Lee, C.; Zhang, G.; Zhang, R.; Vahab, V.; Bong, C. Prevalence and Risk Assessment of Antibiotics in Riverine Estuarine Waters of Larut and Sangga Besar River, Perak. J. Oceanol. Limnol. 2021, 39, 122–134. [Google Scholar] [CrossRef]
  202. Truong, T.; Hoang, T.L.; Tran, L.T.; Pham, T.P.T.; Le, T.-H. Prevalence of Antibiotic Resistance Genes in the Saigon River Impacted by Anthropogenic Activities. Water 2021, 13, 2234. [Google Scholar] [CrossRef]
  203. Giebułtowicz, J.; Nałęcz-Jawecki, G.; Harnisz, M.; Kucharski, D.; Korzeniewska, E.; Płaza, G. Environmental Risk and Risk of Resistance Selection Due to Antimicrobials’ Occurrence in Two Polish Wastewater Treatment Plants and Receiving Surface Water. Molecules 2020, 25, 1470. [Google Scholar] [CrossRef]
  204. Serra-Compte, A.; Pikkemaat, M.G.; Elferink, A.; Almeida, D.; Diogène, J.; Campillo, J.A.; Llorca, M.; Álvarez-Muñoz, D.; Barceló, D.; Rodríguez-Mozaz, S. Combining an Effect-Based Methodology with Chemical Analysis for Antibiotics Determination in Wastewater and Receiving Freshwater and Marine Environment. Environ. Pollut. 2021, 271, 116313. [Google Scholar] [CrossRef] [PubMed]
  205. Gozzo, S.; Moles, S.; Kińska, K.; Ormad, M.P.; Mosteo, R.; Gómez, J.; Laborda, F.; Szpunar, J. Screening for Antibiotics and Their Degradation Products in Surface and Wastewaters of the POCTEFA Territory by Solid-Phase Extraction-UPLC-Electrospray MS/MS. Water 2022, 15, 14. [Google Scholar] [CrossRef]
  206. Wang, L.; Li, H.; Dang, J.; Guo, H.; Zhu, Y.; Han, W. Occurrence, Distribution, and Partitioning of Antibiotics in Surface Water and Sediment in a Typical Tributary of Yellow River, China. Environ. Sci. Pollut. Res. 2021, 28, 28207–28221. [Google Scholar] [CrossRef]
  207. Dong, J.; Yan, D.; Mo, K.; Chen, Q.; Zhang, J.; Chen, Y.; Wang, Z. Antibiotics along an Alpine River and in the Receiving Lake with a Catchment Dominated by Grazing Husbandry. J. Environ. Sci. 2022, 115, 374–382. [Google Scholar] [CrossRef]
  208. Liu, Y.; Cai, D.; Li, X.; Wu, Q.; Ding, P.; Shen, L.; Yang, J.; Hu, G.; Wu, J.; Zhang, L. Occurrence, Fate, and Risk Assessment of Antibiotics in Typical Pharmaceutical Manufactories and Receiving Water Bodies from Different Regions. PLoS ONE 2023, 18, e0270945. [Google Scholar] [CrossRef]
  209. Liu, Y.; Chen, Y.; Feng, M.; Chen, J.; Shen, W.; Zhang, S. Occurrence of Antibiotics and Antibiotic Resistance Genes and Their Correlations in River-Type Drinking Water Source, China. Environ. Sci. Pollut. Res. 2021, 28, 42339–42352. [Google Scholar] [CrossRef] [PubMed]
  210. Lin, H.; Huang, L.; Gao, Z.; Lin, W.; Ren, Y. Comparative Analysis of the Removal and Transformation of 10 Typical Pharmaceutical and Personal Care Products in Secondary Treatment of Sewage: A Case Study of Two Biological Treatment Processes. J. Environ. Chem. Eng. 2022, 10, 107638. [Google Scholar] [CrossRef]
  211. Hanna, N.; Purohit, M.; Diwan, V.; Chandran, S.P.; Riggi, E.; Parashar, V.; Tamhankar, A.J.; Lundborg, C.S. Monitoring of Water Quality, Antibiotic Residues, and Antibiotic-Resistant Escherichia coli in the Kshipra River in India over a 3-Year Period. Int. J. Environ. Res. Public. Health 2020, 17, 7706. [Google Scholar] [CrossRef]
  212. Lenart-Boroń, A.; Prajsnar, J.; Guzik, M.; Boroń, P.; Chmiel, M. How Much of Antibiotics Can Enter Surface Water with Treated Wastewater and How It Affects the Resistance of Waterborne Bacteria: A Case Study of the Białka River Sewage Treatment Plant. Environ. Res. 2020, 191, 110037. [Google Scholar] [CrossRef]
  213. Kolpin, D.; Skopec, M.; Meyer, M.; Furlong, E.; Zaugg, S. Urban Contribution of Pharmaceuticals and Other Organic Wastewater Contaminants to Streams during Differing Flow Conditions. Sci. Total Environ. 2004, 328, 119–130. [Google Scholar] [CrossRef]
  214. Mai, Z.; Xiong, X.; Hu, H.; Jia, J.; Wu, C.; Wang, G. Occurrence, Distribution, and Ecological Risks of Antibiotics in Honghu Lake and Surrounding Aquaculture Ponds, China. Environ. Sci. Pollut. Res. 2023, 30, 50732–50742. [Google Scholar] [CrossRef]
  215. Gray, A.D.; Todd, D.; Hershey, A.E. The Seasonal Distribution and Concentration of Antibiotics in Rural Streams and Drinking Wells in the Piedmont of North Carolina. Sci. Total Environ. 2020, 710, 136286. [Google Scholar] [CrossRef]
  216. Gray, A. The Use of Non-Target High-Resolution Mass Spectrometry Screening to Detect the Presence of Antibiotic Residues in Urban Streams of Greensboro North Carolina. J. Environ. Health Sci. Eng. 2021, 19, 1313–1321. [Google Scholar] [CrossRef]
  217. Nakata, H.; Kannan, K.; Jones, P.D.; Giesy, J.P. Determination of Fluoroquinolone Antibiotics in Wastewater Effluents by Liquid Chromatography–Mass Spectrometry and Fluorescence Detection. Chemosphere 2005, 58, 759–766. [Google Scholar] [CrossRef]
  218. Chen, C.X.; Aris, A.; Yong, E.L.; Noor, Z.Z. Evaluation of the Occurrence of Antibiotics at Different Treatment Stages of Decentralised and Conventional Sewage Treatment Plants. Int. J. Environ. Sci. Technol. 2022, 19, 5547–5562. [Google Scholar] [CrossRef]
  219. Jia, D.; You, X.; Tang, M.; Lyu, Y.; Hu, J.; Sun, W. Single and Combined Genotoxicity of Metals and Fluoroquinolones to Zebrafish Embryos at Environmentally Relevant Concentrations. Aquat. Toxicol. 2023, 258, 106495. [Google Scholar] [CrossRef]
  220. Robinson, D.I. Control of Genotoxic Impurities in Active Pharmaceutical Ingredients: A Review and Perspective. Org. Process Res. Dev. 2010, 14, 946–959. [Google Scholar] [CrossRef]
  221. Turkez, H.; Arslan, M.E.; Ozdemir, O. Genotoxicity Testing: Progress and Prospects for the next Decade. Expert Opin. Drug Metab. Toxicol. 2017, 13, 1089–1098. [Google Scholar] [CrossRef] [PubMed]
  222. Bhattacharya, P.; Mukherjee, S.; Mandal, S.M. Fluoroquinolone Antibiotics Show Genotoxic Effect through DNA-Binding and Oxidative Damage. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2020, 227, 117634. [Google Scholar] [CrossRef] [PubMed]
  223. Thomé, S.; Bizarro, C.R.; Lehmann, M.; Abreu, B.R.R.D.; Andrade, H.H.R.D.; Cunha, K.S.; Dihl, R.R. Recombinagenic and Mutagenic Activities of Fluoroquinolones in Drosophila melanogaster. Mutat. Res. Toxicol. Environ. Mutagen. 2012, 742, 43–47. [Google Scholar] [CrossRef] [PubMed]
  224. Xu, Y.-H.; Wei, X.-L.; Xu, Y.-C.; Zhang, D.-G.; Zhao, T.; Zheng, H.; Luo, Z. Waterborne Enrofloxacin Exposure Activated Oxidative Stress and MAPK Pathway, Induced Apoptosis and Resulted in Immune Dysfunction in the Gills of Yellow Catfish Pelteobagrus fulvidraco. Aquaculture 2022, 547, 737541. [Google Scholar] [CrossRef]
  225. Nunes, B.; Leal, C.; Rodrigues, S.; Antunes, S.C. Assessment of Ecotoxicological Effects of Ciprofloxacin in Daphnia magna: Life-History Traits, Biochemical and Genotoxic Effects. Water Sci. Technol. 2018, 2017, 835–844. [Google Scholar] [CrossRef]
  226. Domagala, J.M.; Hagen, S.E. Structure-Activity Relationships of the Quinolone Antibacterials in the New Millennium: Some Things Change and Some Do Not. In Quinolone Antimicrobial Agents; Hooper, D.C., Rubinstein, E., Eds.; ASM Press: Washington, DC, USA, 2014; pp. 1–18. ISBN 978-1-68367-220-3. [Google Scholar]
  227. Pham, T.D.M.; Ziora, Z.M.; Blaskovich, M.A.T. Quinolone Antibiotics. MedChemComm 2019, 10, 1719–1739. [Google Scholar] [CrossRef]
  228. Fengxian, C.; Reti, H. Analysis of Positions and Substituents on Genotoxicity of Fluoroquinolones with Quantitative Structure-Activity Relationship and 3D Pharmacophore Model. Ecotoxicol. Environ. Saf. 2017, 136, 111–118. [Google Scholar] [CrossRef]
  229. Menon, A.; Pettinari, L.; Martinelli, C.; Colombo, G.; Portinaro, N.; Dalle-Donne, I.; d’Agostino, M.C.; Gagliano, N. New Insights in Extracellular Matrix Remodeling and Collagen Turnover Related Pathways in Cultured Human Tenocytes after Ciprofloxacin Administration. Muscles Ligaments Tendons J. 2013, 3, 122–131. [Google Scholar]
  230. Guzzardi, D.G.; Teng, G.; Kang, S.; Geeraert, P.J.; Pattar, S.S.; Svystonyuk, D.A.; Belke, D.D.; Fedak, P.W.M. Induction of Human Aortic Myofibroblast-Mediated Extracellular Matrix Dysregulation: A Potential Mechanism of Fluoroquinolone-Associated Aortopathy. J. Thorac. Cardiovasc. Surg. 2019, 157, 109–119.e2. [Google Scholar] [CrossRef]
  231. Bennett, A.C.; Bennett, C.L.; Witherspoon, B.J.; Knopf, K.B. An Evaluation of Reports of Ciprofloxacin, Levofloxacin, and Moxifloxacin-Association Neuropsychiatric Toxicities, Long-Term Disability, and Aortic Aneurysms/Dissections Disseminated by the Food and Drug Administration and the European Medicines Agency. Expert Opin. Drug Saf. 2019, 18, 1055–1063. [Google Scholar] [CrossRef]
  232. Orobello, N.C.; Dirain, C.O.; Schultz, G.; Milne-Davies, B.A.; Ng, M.R.A.; Antonelli, P.J. Ciprofloxacin Decreases Collagen in Mouse Tympanic Membrane Fibroblasts. Otolaryngol. Neck Surg. 2016, 155, 127–132. [Google Scholar] [CrossRef] [PubMed]
  233. Sendzik, J.; Shakibaei, M.; Schäfer-Korting, M.; Lode, H.; Stahlmann, R. Synergistic Effects of Dexamethasone and Quinolones on Human-Derived Tendon Cells. Int. J. Antimicrob. Agents 2010, 35, 366–374. [Google Scholar] [CrossRef] [PubMed]
  234. Etminan, M.; Forooghian, F.; Brophy, J.M.; Bird, S.T.; Maberley, D. Oral Fluoroquinolones and the Risk of Retinal Detachment. JAMA 2012, 307, 1414. [Google Scholar] [CrossRef] [PubMed]
  235. Raguideau, F.; Lemaitre, M.; Dray-Spira, R.; Zureik, M. Association Between Oral Fluoroquinolone Use and Retinal Detachment. JAMA Ophthalmol. 2016, 134, 415. [Google Scholar] [CrossRef] [PubMed]
  236. Duan, L.; Deng, H.; Zhang, Q.; Gao, P.; Wu, L. Environmentally Relevant Levels of BPA and NOR Disturb Early Skeletal Development in Zebrafish. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2023, 271, 109662. [Google Scholar] [CrossRef] [PubMed]
  237. Jia, D.; Zhang, R.; Shao, J.; Zhang, W.; Cai, L.; Sun, W. Exposure to Trace Levels of Metals and Fluoroquinolones Increases Inflammation and Tumorigenesis Risk of Zebrafish Embryos. Environ. Sci. Ecotechnology 2022, 10, 100162. [Google Scholar] [CrossRef] [PubMed]
  238. Akahane, K.; Sekiguchi, M.; Une, T.; Osada, Y. Structure-Epileptogenicity Relationship of Quinolones with Special Reference to Their Interaction with Gamma-Aminobutyric Acid Receptor Sites. Antimicrob. Agents Chemother. 1989, 33, 1704–1708. [Google Scholar] [CrossRef] [PubMed]
  239. Xiao, C.; Han, Y.; Liu, Y.; Zhang, J.; Hu, C. Relationship Between Fluoroquinolone Structure and Neurotoxicity Revealed by Zebrafish Neurobehavior. Chem. Res. Toxicol. 2018, 31, 238–250. [Google Scholar] [CrossRef] [PubMed]
  240. Mehlhorn, A.J.; Brown, D.A. Safety Concerns with Fluoroquinolones. Ann. Pharmacother. 2007, 41, 1859–1866. [Google Scholar] [CrossRef] [PubMed]
  241. Rubio-Nazabal, E.; Álvarez-Pérez, P.; Sánchez-Vidal, E. Síndrome confusional agudo por levofloxacino. Enferm. Infecc. Microbiol. Clínica 2013, 31, 704–705. [Google Scholar] [CrossRef]
  242. Xi, J.; Liu, J.; He, S.; Shen, W.; Wei, C.; Li, K.; Zhang, Y.; Yue, J.; Yang, Z. Effects of Norfloxacin Exposure on Neurodevelopment of Zebrafish (Danio rerio) Embryos. NeuroToxicology 2019, 72, 85–94. [Google Scholar] [CrossRef] [PubMed]
  243. Ebert, I.; Bachmann, J.; Kühnen, U.; Küster, A.; Kussatz, C.; Maletzki, D.; Schlüter, C. Toxicity of the Fluoroquinolone Antibiotics Enrofloxacin and Ciprofloxacin to Photoautotrophic Aquatic Organisms. Environ. Toxicol. Chem. 2011, 30, 2786–2792. [Google Scholar] [CrossRef]
  244. González-Pleiter, M.; Gonzalo, S.; Rodea-Palomares, I.; Leganés, F.; Rosal, R.; Boltes, K.; Marco, E.; Fernández-Piñas, 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]
  245. Robinson, A.A.; Belden, J.B.; Lydy, M.J. Toxicity of Fluoroquinolone Antibiotics to Aquatic Organisms. Environ. Toxicol. Chem. 2005, 24, 423–430. [Google Scholar] [CrossRef]
  246. Halling-Sorensen, B.; Lützhøft, H.; Andersen, H.R.; Ingerslev, F. Environmental Risk Assessment of Antibiotics: Comparison of Mecillinam, Trimethoprim and Ciprofloxacin. J. Antimicrob. Chemother. 2000, 46, 53–58. [Google Scholar] [CrossRef]
  247. Lützhøft, H.-C.H.; Halling-Sørensen, B.; Jørgensen, S.E. Algal Toxicity of Antibacterial Agents Applied in Danish Fish Farming. Arch. Environ. Contam. Toxicol. 1999, 36, 1–6. [Google Scholar] [CrossRef]
  248. Wan, L.; Wu, Y.; Zhang, B.; Yang, W.; Ding, H.; Zhang, W. Effects of Moxifloxacin and Gatifloxacin Stress on Growth, Photosynthesis, Antioxidant Responses, and Microcystin Release in Microcystis aeruginosa. J. Hazard. Mater. 2021, 409, 124518. [Google Scholar] [CrossRef]
  249. Zhao, L.; Xu, K.; Juneau, P.; Huang, P.; Lian, Y.; Zheng, X.; Zhong, Q.; Zhang, W.; Xiao, F.; Wu, B.; et al. Light Modulates the Effect of Antibiotic Norfloxacin on Photosynthetic Processes of Microcystis aeruginosa. Aquat. Toxicol. 2021, 235, 105826. [Google Scholar] [CrossRef]
  250. Magdaleno, A.; Saenz, M.E.; Juárez, A.B.; Moretton, J. Effects of Six Antibiotics and Their Binary Mixtures on Growth of Pseudokirchneriella subcapitata. Ecotoxicol. Environ. Saf. 2015, 113, 72–78. [Google Scholar] [CrossRef]
  251. Martins, N.; Pereira, R.; Abrantes, N.; Pereira, J.; Gonçalves, F.; Marques, C.R. Ecotoxicological Effects of Ciprofloxacin on Freshwater Species: Data Integration and Derivation of Toxicity Thresholds for Risk Assessment. Ecotoxicology 2012, 21, 1167–1176. [Google Scholar] [CrossRef]
  252. Yang, L.; Ying, G.; Su, H.; Stauber, J.L.; Adams, M.S.; Binet, M.T. Growth-inhibiting Effects of 12 Antibacterial Agents and Their Mixtures on the Freshwater Microalga Pseudokirchneriella subcapitata. Environ. Toxicol. Chem. 2008, 27, 1201–1208. [Google Scholar] [CrossRef]
  253. Fu, L.; Huang, T.; Wang, S.; Wang, X.; Su, L.; Li, C.; Zhao, Y. Toxicity of 13 Different Antibiotics towards Freshwater Green Algae Pseudokirchneriella subcapitata and Their Modes of Action. Chemosphere 2017, 168, 217–222. [Google Scholar] [CrossRef]
  254. Zounková, R.; Klimešová, Z.; Nepejchalová, L.; Hilscherová, K.; Bláha, L. Complex Evaluation of Ecotoxicity and Genotoxicity of Antimicrobials Oxytetracycline and Flumequine Used in Aquaculture. Environ. Toxicol. Chem. 2011, 30, 1184–1189. [Google Scholar] [CrossRef]
  255. Van Der Grinten, E.; Pikkemaat, M.G.; Van Den Brandhof, E.-J.; Stroomberg, G.J.; Kraak, M.H.S. Comparing the Sensitivity of Algal, Cyanobacterial and Bacterial Bioassays to Different Groups of Antibiotics. Chemosphere 2010, 80, 1–6. [Google Scholar] [CrossRef]
  256. Christensen, A.M.; Ingerslev, F.; Baun, A. Ecotoxicity of Mixtures of Antibiotics Used in Aquacultures. Environ. Toxicol. Chem. 2006, 25, 2208–2215. [Google Scholar] [CrossRef]
  257. Yamashita, N.; Yasojima, M.; Nakada, N.; Miyajima, K.; Komori, K.; Suzuki, Y.; Tanaka, H. Effects of Antibacterial Agents, Levofloxacin and Clarithromycin, on Aquatic Organisms. Water Sci. Technol. 2006, 53, 65–72. [Google Scholar] [CrossRef]
  258. Eguchi, K.; Nagase, H.; Ozawa, M.; Endoh, Y.S.; Goto, K.; Hirata, K.; Miyamoto, K.; Yoshimura, H. Evaluation of Antimicrobial Agents for Veterinary Use in the Ecotoxicity Test Using Microalgae. Chemosphere 2004, 57, 1733–1738. [Google Scholar] [CrossRef]
  259. Isidori, M.; Lavorgna, M.; Nardelli, A.; Pascarella, L.; Parrella, A. Toxic and Genotoxic Evaluation of Six Antibiotics on Non-Target Organisms. Sci. Total Environ. 2005, 346, 87–98. [Google Scholar] [CrossRef]
  260. Ferrari, B.; Mons, R.; Vollat, B.; Fraysse, B.; Paxēaus, N.; Giudice, R.L.; Pollio, A.; Garric, J. Environmental Risk Assessment of Six Human Pharmaceuticals: Are the Current Environmental Risk Assessment Procedures Sufficient for the Protection of the Aquatic Environment? Environ. Toxicol. Chem. 2004, 23, 1344–1354. [Google Scholar] [CrossRef]
  261. Wan, L.; Wu, Y.; Zhang, Y.; Zhang, W. Toxicity, Biodegradation of Moxifloxacin and Gatifloxacin on Chlamydomonas reinhardtii and Their Metabolic Fate. Ecotoxicol. Environ. Saf. 2022, 240, 113711. [Google Scholar] [CrossRef]
  262. Geiger, E.; Hornek-Gausterer, R.; Saçan, M.T. Single and Mixture Toxicity of Pharmaceuticals and Chlorophenols to Freshwater Algae Chlorella vulgaris. Ecotoxicol. Environ. Saf. 2016, 129, 189–198. [Google Scholar] [CrossRef]
  263. Nie, X.; Wang, X.; Chen, J.; Zitko, V.; An, T. Response of the Freshwater Alga Chlorella vulgaris to Trichloroisocyanuric Acid and Ciprofloxacin. Environ. Toxicol. Chem. 2008, 27, 168–173. [Google Scholar] [CrossRef]
  264. Li, Z.; Gao, X.; Bao, J.; Li, S.; Wang, X.; Li, Z.; Zhu, L. Evaluation of Growth and Antioxidant Responses of Freshwater Microalgae Chlorella sorokiniana and Scenedesmus dimorphus under Exposure of Moxifloxacin. Sci. Total Environ. 2023, 858, 159788. [Google Scholar] [CrossRef]
  265. Backhaus, T.; Faust, M.; Junghans, M.; Scholze, M.; Grimme, H. Low Algal Toxicities of Quinolones Confirm Their Specific Molecular Mechanism of Action. In Proceedings of the 11th Annual Meeting of SETAC Europe, Madrid, Spain, 6–10 May 2001. [Google Scholar]
  266. Qin, H.; Chen, L.; Lu, N.; Zhao, Y.; Yuan, X. Toxic Effects of Enrofloxacin on Scenedesmus obliquus. Front. Environ. Sci. Eng. 2012, 6, 107–116. [Google Scholar] [CrossRef]
  267. Nie, X.; Gu, J.; Lu, J.; Pan, W.; Yang, Y. Effects of Norfloxacin and Butylated Hydroxyanisole on the Freshwater Microalga Scenedesmus obliquus. Ecotoxicology 2009, 18, 677–684. [Google Scholar] [CrossRef]
  268. Lu, J.Y.; Li, X.; Yang, Y.T.; Nie, X.P. Toxic effects of nutylated hydroxyanisole and norfloxacin on aquatic organisms. Ecol. Sci. 2007, 26, 55–58. [Google Scholar]
  269. Brain, R.A.; Johnson, D.J.; Richards, S.M.; Sanderson, H.; Sibley, P.K.; Solomon, K.R. Effects of 25 Pharmaceutical Compounds to Lemna gibba Using a Seven-day Static-renewal Test. Environ. Toxicol. Chem. 2004, 23, 371–382. [Google Scholar] [CrossRef]
  270. Brooks, B.W.; Maul, J.D.; Belden, J.B. Antibiotics in Aquatic and Terrestrial Ecosystems. In Encyclopedia of Ecology; Elsevier: Amsterdam, The Netherlands, 2008; pp. 210–217. ISBN 978-0-08-045405-4. [Google Scholar]
  271. Eluk, D.; Nagel, O.; Gagneten, A.; Reno, U.; Althaus, R. Toxicity of Fluoroquinolones on the Cladoceran Daphnia magna. Water Environ. Res. 2021, 93, 2914–2930. [Google Scholar] [CrossRef]
  272. Dionísio, R.; Daniel, D.; Alkimin, G.D.D.; Nunes, B. Multi-Parametric Analysis of Ciprofloxacin Toxicity at Ecologically Relevant Levels: Short- and Long-Term Effects on Daphnia magna. Environ. Toxicol. Pharmacol. 2020, 74, 103295. [Google Scholar] [CrossRef]
  273. Dalla Bona, M.; Di Leva, V.; De Liguoro, M. The Sensitivity of Daphnia magna and Daphnia curvirostris to 10 Veterinary Antibacterials and to Some of Their Binary Mixtures. Chemosphere 2014, 115, 67–74. [Google Scholar] [CrossRef]
  274. Mala, A.A.; Dutta, J. Acute and Chronic Toxicity of Fluoroquinolone Antibiotics through Fresh Water Cladoceron Daphnia magna. Indian J. Ecol. 2019, 46, 874–879. [Google Scholar]
  275. Kergaravat, S.V.; Hernández, S.R.; Gagneten, A.M. Second-, Third- and Fourth-Generation Quinolones: Ecotoxicity Effects on Daphnia and Ceriodaphnia Species. Chemosphere 2021, 262, 127823. [Google Scholar] [CrossRef] [PubMed]
  276. Luo, T.; Chen, J.; Li, X.; Zhang, S.; Yao, H.; Peijnenburg, W.J.G.M. Effects of Lomefloxacin on Survival, Growth and Reproduction of Daphnia magna under Simulated Sunlight Radiation. Ecotoxicol. Environ. Saf. 2018, 166, 63–70. [Google Scholar] [CrossRef] [PubMed]
  277. Han, Y.; Ma, Y.; Yao, S.; Zhang, J.; Hu, C. In Vivo and in Silico Evaluations of Survival and Cardiac Developmental Toxicity of Quinolone Antibiotics in Zebrafish Embryos (Danio rerio). Environ. Pollut. 2021, 277, 116779. [Google Scholar] [CrossRef] [PubMed]
  278. Özhan Turhan, D. Evaluation of Teratogenic and Toxic Effects of Enrofloxacin-Based Antibiotic on Zebrafish (Danio rerio) Larvae with Biochemical and Developmental Markers. Chem. Ecol. 2021, 37, 704–714. [Google Scholar] [CrossRef]
  279. Lancieri, M.; Cozzolino, S.; Filosa, S.; Motta, C.M.; Tammaro, S.; Gaudio, L.; Migliore, L. Toxicity and Teratogenic Effects of Flumequine on Danio rerio Embryos. Fresenius Environ. Bullettin 2002, 11, 642–646. [Google Scholar]
  280. Sharma, L.; Siedlewicz, G.; Pazdro, K. The Toxic Effects of Antibiotics on Freshwater and Marine Photosynthetic Microorganisms: State of the Art. Plants 2021, 10, 591. [Google Scholar] [CrossRef]
  281. Aderemi, A.O.; Novais, S.C.; Lemos, M.F.L.; Alves, L.M.; Hunter, C.; Pahl, O. Oxidative Stress Responses and Cellular Energy Allocation Changes in Microalgae Following Exposure to Widely Used Human Antibiotics. Aquat. Toxicol. 2018, 203, 130–139. [Google Scholar] [CrossRef] [PubMed]
  282. Mater, N.; Geret, F.; Castillo, L.; Faucet-Marquis, V.; Albasi, C.; Pfohl-Leszkowicz, A. In Vitro Tests Aiding Ecological Risk Assessment of Ciprofloxacin, Tamoxifen and Cyclophosphamide in Range of Concentrations Released in Hospital Wastewater and Surface Water. Environ. Int. 2014, 63, 191–200. [Google Scholar] [CrossRef]
  283. Liu, B.; Nie, X.; Liu, W.; Snoeijs, P.; Guan, C.; Tsui, M.T.K. Toxic Effects of Erythromycin, Ciprofloxacin and Sulfamethoxazole on Photosynthetic Apparatus in Selenastrum capricornutum. Ecotoxicol. Environ. Saf. 2011, 74, 1027–1035. [Google Scholar] [CrossRef]
  284. Henderson, K.L.; Coats, J.R. (Eds.) Veterinary Pharmaceuticals in the Environment; ACS Symposium Series; American Chemical Society: Washington, DC, USA, 2010; Volume 1018, ISBN 978-0-8412-6962-0. [Google Scholar]
  285. Migliore, L.; Cozzolino, S.; Fiori, M. Phytotoxicity to and Uptake of Flumequine Used in Intensive Aquaculture on the Aquatic Weed, Lythrum salicaria L. Chemosphere 2000, 40, 741–750. [Google Scholar] [CrossRef]
  286. Nunes, B.; Veiga, V.; Frankenbach, S.; Serôdio, J.; Pinto, G. Evaluation of Physiological Changes Induced by the Fluoroquinolone Antibiotic Ciprofloxacin in the Freshwater Macrophyte Species Lemna minor and Lemna gibba. Environ. Toxicol. Pharmacol. 2019, 72, 103242. [Google Scholar] [CrossRef]
  287. Weber, K.P.; Mitzel, M.R.; Slawson, R.M.; Legge, R.L. Effect of Ciprofloxacin on Microbiological Development in Wetland Mesocosms. Water Res. 2011, 45, 3185–3196. [Google Scholar] [CrossRef]
  288. Pan, Y.; Yan, S.; Li, R.; Hu, Y.; Chang, X. Lethal/Sublethal Responses of Daphnia magna to Acute Norfloxacin Contamination and Changes in Phytoplankton-Zooplankton Interactions Induced by This Antibiotic. Sci. Rep. 2017, 7, 40385. [Google Scholar] [CrossRef] [PubMed]
  289. Dalla Bona, M.; Lizzi, F.; Borgato, A.; De Liguoro, M. Increasing Toxicity of Enrofloxacin over Four Generations of Daphnia magna. Ecotoxicol. Environ. Saf. 2016, 132, 397–402. [Google Scholar] [CrossRef]
  290. Zhang, Y.; Wang, L.; Zhuang, H.; Li, X.; Gao, X.; An, Z.; Liu, X.; Yang, H.; Wei, W.; Zhang, X. Excessive Use of Enrofloxacin Leads to Growth Inhibition of Juvenile Giant Freshwater Prawn Macrobrachium rosenbergii. Ecotoxicol. Environ. Saf. 2019, 169, 344–352. [Google Scholar] [CrossRef]
  291. Kim, J.-W.; Ishibashi, H.; Yamauchi, R.; Ichikawa, N.; Takao, Y.; Hirano, M.; Koga, M.; Arizono, K. Acute Toxicity of Pharmaceutical and Personal Care Products on Freshwater Crustacean (Thamnocephalus platyurus) and Fish (Oryzias latipes). J. Toxicol. Sci. 2009, 34, 227–232. [Google Scholar] [CrossRef] [PubMed]
  292. Pietropoli, E.; Pauletto, M.; Iori, S.; Tolosi, R.; Giantin, M.; Dacasto, M.; De Liguoro, M. Transgenerational Effects of the Fluoroquinolone Flumequine in D. Magna: An In Vivo Whole-Transcriptomic Investigation. In Proceedings of the 34th Annual Meeting of SETAC Europe, Seville, Spain, 5–9 May 2024. [Google Scholar]
  293. Jager, T. Identifying and Predicting Delayed Mortality with Toxicokinetic–Toxicodynamic Models. Environ. Toxicol. Chem. 2024, 43, 1030–1035. [Google Scholar] [CrossRef]
  294. Shen, R.; Yu, Y.; Lan, R.; Yu, R.; Yuan, Z.; Xia, Z. The Cardiovascular Toxicity Induced by High Doses of Gatifloxacin and Ciprofloxacin in Zebrafish. Environ. Pollut. 2019, 254, 112861. [Google Scholar] [CrossRef]
  295. Nogueira, A.F.; Pinto, G.; Correia, B.; Nunes, B. Embryonic Development, Locomotor Behavior, Biochemical, and Epigenetic Effects of the Pharmaceutical Drugs Paracetamol and Ciprofloxacin in Larvae and Embryos of Danio rerio When Exposed to Environmental Realistic Levels of Both Drugs. Environ. Toxicol. 2019, 34, 1177–1190. [Google Scholar] [CrossRef]
  296. Liu, J.; Lu, G.; Wu, D.; Yan, Z. A Multi-Biomarker Assessment of Single and Combined Effects of Norfloxacin and Sulfamethoxazole on Male Goldfish (Carassius auratus). Ecotoxicol. Environ. Saf. 2014, 102, 12–17. [Google Scholar] [CrossRef]
  297. Liang, X.; Wang, L.; Ou, R.; Nie, X.; Yang, Y.; Wang, F.; Li, K. Effects of Norfloxacin on Hepatic Genes Expression of P450 Isoforms (CYP1A and CYP3A), GST and P-Glycoprotein (P-Gp) in Swordtail Fish (Xiphophorus helleri). Ecotoxicology 2015, 24, 1566–1573. [Google Scholar] [CrossRef]
  298. Liang, X.; Wang, F.; Li, K.; Nie, X.; Fang, H. Effects of Norfloxacin Nicotinate on the Early Life Stage of Zebrafish (Danio rerio): Developmental Toxicity, Oxidative Stress and Immunotoxicity. Fish Shellfish. Immunol. 2020, 96, 262–269. [Google Scholar] [CrossRef] [PubMed]
  299. Zhao, X.-L.; Li, P.; Zhang, S.-Q.; He, S.-W.; Xing, S.-Y.; Cao, Z.-H.; Lu, R.; Li, Z.-H. Effects of Environmental Norfloxacin Concentrations on the Intestinal Health and Function of Juvenile Common Carp and Potential Risk to Humans. Environ. Pollut. 2021, 287, 117612. [Google Scholar] [CrossRef] [PubMed]
  300. Yang, X.; Xu, X.; Wei, X.; Wan, J.; Zhang, Y. Biomarker Effects in Carassius auratus Exposure to Ofloxacin, Sulfamethoxazole and Ibuprofen. Int. J. Environ. Res. Public. Health 2019, 16, 1628. [Google Scholar] [CrossRef] [PubMed]
  301. Lee, S.; Kim, C.; Liu, X.; Lee, S.; Kho, Y.; Kim, W.-K.; Kim, P.; Choi, K. Ecological Risk Assessment of Amoxicillin, Enrofloxacin, and Neomycin: Are Their Current Levels in the Freshwater Environment Safe? Toxics 2021, 9, 196. [Google Scholar] [CrossRef] [PubMed]
  302. Park, S.; Choi, K. Hazard Assessment of Commonly Used Agricultural Antibiotics on Aquatic Ecosystems. Ecotoxicology 2008, 17, 526–538. [Google Scholar] [CrossRef] [PubMed]
  303. Zhang, Y.; Wang, X.; Yin, X.; Shi, M.; Dahlgren, R.A.; Wang, H. Toxicity Assessment of Combined Fluoroquinolone and Tetracycline Exposure in Zebrafish (Danio rerio): Toxicology Assessment of Fluoroquinolone and Tetracycline in Zebrafish. Environ. Toxicol. 2016, 31, 736–750. [Google Scholar] [CrossRef] [PubMed]
  304. He, S.; Sun, Q.; Liu, Y.; Li, L.; Yang, X. Acute Toxicity, Bioaccumulation, and Bioreduction of Two Antibiotics, Norfloxacin and Ofloxacin, in the Snail Bellamya aeruginosa. Water. Air. Soil Pollut. 2020, 231, 96. [Google Scholar] [CrossRef]
  305. Gilroy, È.A.M.; Klinck, J.S.; Campbell, S.D.; McInnis, R.; Gillis, P.L.; De Solla, S.R. Toxicity and Bioconcentration of the Pharmaceuticals Moxifloxacin, Rosuvastatin, and Drospirenone to the Unionid Mussel Lampsilis siliquoidea. Sci. Total Environ. 2014, 487, 537–544. [Google Scholar] [CrossRef] [PubMed]
  306. Nentwig, G. Another Example of Effects of Pharmaceuticals on Aquatic Invertebrates: Fluoxetine and Ciprofloxacin. In Pharmaceuticals in the Environment; Kümmerer, K., Ed.; Springer: Berlin/Heidelberg, Germany, 2008; pp. 205–222. ISBN 978-3-540-74663-8. [Google Scholar]
  307. Richards, S.M.; Cole, S.E. A Toxicity and Hazard Assessment of Fourteen Pharmaceuticals to Xenopus laevis Larvae. Ecotoxicology 2006, 15, 647–656. [Google Scholar] [CrossRef]
  308. Giddings, J.M.; Wirtz, J.; Campana, D.; Dobbs, M. Derivation of Combined Species Sensitivity Distributions for Acute Toxicity of Pyrethroids to Aquatic Animals. Ecotoxicology 2019, 28, 242–250. [Google Scholar] [CrossRef]
  309. USEPA SSD Generator V1. Available online: https://www.epa.gov/caddis/download-software (accessed on 15 May 2024).
  310. Nie, X.P.; Lu, J.Y.; Li, X.; Yang, Y.F. Toxic Effects of Norfloxacin to the Growth and the Activity of Antioxidase of Chlorella pyrenoidosa. Asian J. Ecotoxicol. 2007, 2, 327–332. [Google Scholar]
  311. Spurgeon, D.; Lahive, E.; Robinson, A.; Short, S.; Kille, P. Species Sensitivity to Toxic Substances: Evolution, Ecology and Applications. Front. Environ. Sci. 2020, 8, 588380. [Google Scholar] [CrossRef]
  312. Sorgog, K.; Kamo, M. Quantifying the Precision of Ecological Risk: Conventional Assessment Factor Method vs. Species Sensitivity Distribution Method. Ecotoxicol. Environ. Saf. 2019, 183, 109494. [Google Scholar] [CrossRef] [PubMed]
  313. European Chemicals Agency. Guidance on Information Requirements and Chemical Safety Assessment. Chapter R.10: Characterisation of Dose [Concentration]-Response for Environment; European Chemicals Agency: Helsinki, Finland, 2008. [Google Scholar]
  314. OECD Test No. 203: Fish, Acute Toxicity Test; OECD Guidelines for the Testing of Chemicals; Section 2; OECD: Paris, France, 2019; ISBN 978-92-64-06996-1.
  315. European Medicines Agency’s Committee for Medicinal Products for Veterinary Use (CVMP). Guideline on Environmental Impact Assessment for Veterinary Medicinal Products in Support of the VICH Guidelines GL6 and GL38; 2005. Available online: https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-environmental-impact-assessment-veterinary-medicinal-products-support-vich-guidelines-gl6-and-gl38_en.pdf (accessed on 15 May 2024).
  316. Lulijwa, R.; Rupia, E.J.; Alfaro, A.C. Antibiotic Use in Aquaculture, Policies and Regulation, Health and Environmental Risks: A Review of the Top 15 Major Producers. Rev. Aquac. 2020, 12, 640–663. [Google Scholar] [CrossRef]
  317. Al-Omar, M.A. Ciprofloxacin: Drug Metabolism and Pharmacokinetic Profile. In Profiles of Drug Substances, Excipients and Related Methodology; Elsevier: Amsterdam, The Netherlands, 2005; Volume 31, pp. 209–214. ISBN 978-0-12-260831-5. [Google Scholar]
  318. Sukul, P.; Lamshöft, M.; Kusari, S.; Zühlke, S.; Spiteller, M. Metabolism and Excretion Kinetics of 14C-Labeled and Non-Labeled Difloxacin in Pigs after Oral Administration, and Antimicrobial Activity of Manure Containing Difloxacin and Its Metabolites. Environ. Res. 2009, 109, 225–231. [Google Scholar] [CrossRef] [PubMed]
  319. Junza, A.; Barbosa, S.; Codony, M.R.; Jubert, A.; Barbosa, J.; Barrón, D. Identification of Metabolites and Thermal Transformation Products of Quinolones in Raw Cow’s Milk by Liquid Chromatography Coupled to High-Resolution Mass Spectrometry. J. Agric. Food Chem. 2014, 62, 2008–2021. [Google Scholar] [CrossRef] [PubMed]
  320. Rusch, M.; Spielmeyer, A.; Zorn, H.; Hamscher, G. Degradation and Transformation of Fluoroquinolones by Microorganisms with Special Emphasis on Ciprofloxacin. Appl. Microbiol. Biotechnol. 2019, 103, 6933–6948. [Google Scholar] [CrossRef]
  321. Li, Y.; Niu, J.; Wang, W. Photolysis of Enrofloxacin in Aqueous Systems under Simulated Sunlight Irradiation: Kinetics, Mechanism and Toxicity of Photolysis Products. Chemosphere 2011, 85, 892–897. [Google Scholar] [CrossRef] [PubMed]
  322. Loewe, S.; Muischnek, H. Über Kombinationswirkungen: Mitteilung: Hilfsmittel der Fragestellung. Arch. Für Exp. Pathol. Pharmakol. 1926, 114, 313–326. [Google Scholar] [CrossRef]
  323. Backhaus, T.; Scholze, M.; Grimme, L.H. The Single Substance and Mixture Toxicity of Quinolones to the Bioluminescent Bacterium Vibrio fischeri. Aquat. Toxicol. 2000, 49, 49–61. [Google Scholar] [CrossRef]
  324. Cedergreen, N. Quantifying Synergy: A Systematic Review of Mixture Toxicity Studies within Environmental Toxicology. PLoS ONE 2014, 9, e96580. [Google Scholar] [CrossRef] [PubMed]
  325. Khadra, A.; Pinelli, E.; Lacroix, M.Z.; Bousquet-Melou, A.; Hamdi, H.; Merlina, G.; Guiresse, M.; Hafidi, M. Assessment of the Genotoxicity of Quinolone and Fluoroquinolones Contaminated Soil with the Vicia faba Micronucleus Test. Ecotoxicol. Environ. Saf. 2012, 76, 187–192. [Google Scholar] [CrossRef] [PubMed]
  326. Covich, A.P.; Palmer, M.A.; Crowl, T.A. The Role of Benthic Invertebrate Species in Freshwater Ecosystems. BioScience 1999, 49, 119. [Google Scholar] [CrossRef]
Figure 1. Highest levels of fluoroquinolones detected in surface waters (a) and WWTP effluents (b) of Europe, the USA, and Asia. The red spot size indicates the concentration range (see inset). Data are obtained from [24,25,33,40,41,42,46,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218].
Figure 1. Highest levels of fluoroquinolones detected in surface waters (a) and WWTP effluents (b) of Europe, the USA, and Asia. The red spot size indicates the concentration range (see inset). Data are obtained from [24,25,33,40,41,42,46,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218].
Jox 14 00042 g001
Figure 2. EC50 values (mg L−1) of fluoroquinolones in different freshwater taxa. Data are obtained from references listed in Table 1. * compounds that have been withdrawn from the market.
Figure 2. EC50 values (mg L−1) of fluoroquinolones in different freshwater taxa. Data are obtained from references listed in Table 1. * compounds that have been withdrawn from the market.
Jox 14 00042 g002
Figure 3. Species sensitivity distributions for twelve autotroph organisms, based on Microcystis aeruginosa equivalents for eleven fluoroquinolones; circles are equivalents for individual species, the solid line is model-fitted distribution, and greyed lines indicate 95% prediction interval.
Figure 3. Species sensitivity distributions for twelve autotroph organisms, based on Microcystis aeruginosa equivalents for eleven fluoroquinolones; circles are equivalents for individual species, the solid line is model-fitted distribution, and greyed lines indicate 95% prediction interval.
Jox 14 00042 g003
Figure 4. Box plot of the highest concentrations of fluoroquinolones detected in the freshwater environment compared with the PNEC calculated either in the traditional way, by applying a specific Assessment Factor to the lowest EC50 of cyanobacteria, unicellular algae, plants, crustaceans, and fish, or through a normalized Species Sensitivity Distribution in autotroph organisms (AF of 5 applied to the Hazard Concentration fifth percentile—HC5).
Figure 4. Box plot of the highest concentrations of fluoroquinolones detected in the freshwater environment compared with the PNEC calculated either in the traditional way, by applying a specific Assessment Factor to the lowest EC50 of cyanobacteria, unicellular algae, plants, crustaceans, and fish, or through a normalized Species Sensitivity Distribution in autotroph organisms (AF of 5 applied to the Hazard Concentration fifth percentile—HC5).
Jox 14 00042 g004
Table 1. EC50s (mg L−1) reported for FQs in different taxa with their relative reference. Details on test duration, endpoint, and measured parameters are also provided.
Table 1. EC50s (mg L−1) reported for FQs in different taxa with their relative reference. Details on test duration, endpoint, and measured parameters are also provided.
Taxa/SpeciesFluoroquinolonesEC50DurationEndpointParameterReference
Cyanobacteria
Anabaena flos-aquaeciprofloxacin0.010272 hYield inhibitionCells numberEbert et al., 2011 [243]
enrofloxacin0.17372 hYield inhibitionCells numberEbert et al., 2011 [243]
(Anabaena CPB4337)levofloxacin4.872 hReduced luminescenceLuminescenceGonzález-Pleiter et al., 2013 [244]
norfloxacin5.672 hReduced luminescenceLuminescenceGonzález-Pleiter et al., 2013 [244]
Microcystis aeruginosaciprofloxacin0.0175 dGrowth rate inhibitionFluorescenceRobinson et al., 2005 [245]
0.00572 hGrowth rate inhibitionAbsorbanceHalling-Sorensen et al., 2000 [246]
clinafloxacin0.1035 dGrowth rate inhibitionFluorescenceRobinson et al., 2005 [245]
enrofloxacin0.0495 dGrowth rate inhibitionFluorescenceRobinson et al., 2005 [245]
flumequine1.9605 dGrowth rate inhibitionFluorescenceRobinson et al., 2005 [245]
7 dGrowth rate inhibitionFluorescenceLützhøft et al., 1999 [247]
gatifloxacin0.0253096 hGrowth rate inhibitionFluorescenceWan et al., 2021 [248]
levofloxacin0.00795 dGrowth rate inhibitionFluorescenceRobinson et al., 2005 [245]
lomefloxacin0.1865 dGrowth rate inhibitionFluorescenceRobinson et al., 2005 [245]
moxifloxacin0.0603496 hGrowth rate inhibitionFluorescenceWan et al., 2021 [248]
norfloxacin0.0347972 hGrowth rate inhibitionFluorescenceZhao et al., 2021 [249]
ofloxacin0.0215 dGrowth rate inhibitionFluorescenceRobinson et al., 2005 [245]
sarafloxacin0.0157 dGrowth rate inhibitionFluorescenceLützhøft et al., 1999 [247]
Microcystis panniformisciprofloxacin0.0135696 hGrowth rate inhibitionCells numberAzevedo et al., 2019 [171]
Unicellular green algae
Raphidocelis subcapitataciprofloxacin11.372 hGrowth rate inhibitionAbsorbanceMagdaleno et al., 2015 [250]
2.9772 hGrowth rate inhibitionAbsorbanceHalling-Sorensen et al., 2000 [246]
4.8396 hGrowth rate inhibitionCells numberMartins et al., 2012 [251]
18.772 hGrowth rate inhibitionFluorescenceRobinson et al., 2005 [245]
6.772 hYield inhibitionAbsorbanceYang et al., 2008 [252]
7.08296 hGrowth rate inhibitionAbsorbanceFu et al., 2017 [253]
clinafloxacin1.172 hGrowth rate inhibitionFluorescenceRobinson et al., 2005 [245]
enrofloxacin3.172 hGrowth rate inhibitionFluorescenceRobinson et al., 2005 [245]
flumequine572 hGrowth rate inhibitionFluorescenceRobinson et al., 2005 [245]
572 hGrowth rate inhibitionFluorescenceLützhøft et al., 1999 [247]
2.696 hGrowth rate inhibitionAbsorbanceZounková et al., 2011 [254]
1624 hYield inhibitionFluorescenceVan Der Grinten et al., 2010 [255]
8.148 hGrowth rate inhibitionFluorescenceChristensen et al., 2006 [256]
levofloxacin7.472 hGrowth rate inhibitionFluorescenceRobinson et al., 2005 [245]
1.296 hGrowth rate inhibitionAbsorbanceYamashita et al., 2006 [257]
lomefloxacin22.772 hGrowth rate inhibitionFluorescenceRobinson et al., 2005 [245]
norfloxacin16.672 hGrowth rate inhibitionCells countEguchi et al., 2004 [258]
1872 hYield inhibitionAbsorbanceYang et al., 2008 [252]
59.40496 hGrowth rate inhibitionAbsorbanceFu et al., 2017 [253]
ofloxacin12.172 hGrowth rate inhibitionFluorescenceRobinson et al., 2005 [245]
4.24196 hGrowth rate inhibitionAbsorbanceFu et al., 2017 [253]
1.4472 hGrowth rate inhibitionCells numberIsidori et al., 2005 [259]
4.7496 hGrowth rate inhibitionCells numberFerrari et al., 2004 [260]
sarafloxacin1672 hGrowth rate inhibitionFluorescenceLützhøft et al., 1999 [247]
Chlamydomonas reinhardtiigatifloxacin83.047 dGrowth rate inhibitionAbsorbanceWan et al., 2022 [261]
moxifloxacin12.657 dGrowth rate inhibitionAbsorbanceWan et al., 2022 [261]
Chlorella vulgarisciprofloxacin29.0996 hGrowth rate inhibitionAbsorbanceGeiger et al., 2016 [262]
20.696 hGrowth rate inhibitionCells numberNie et al., 2008 [263]
norfloxacin10.472 hGrowth rate inhibitionCells countEguchi et al., 2004 [258]
Chlorella sorokinianamoxifloxacin2896 hGrowth rate inhibitionAbsorbanceLi et al., 2023 [264]
Chlorella sp.ciprofloxacin2372 hGrowth rate inhibitionCells numberAndrieu et al., 2015 [53]
enrofloxacin11172 hGrowth rate inhibitionCells numberAndrieu et al., 2015 [53]
Desmodesmus subspicatusenrofloxacin5.56872 hYield inhibitionCells numberEbert et al., 2011 [243]
Scenedesmus vacuolatusflumequine3.724 hGrowth rate inhibitionCells numberBackhaus et al., 2001 [265]
lomefloxacin5824 hGrowth rate inhibitionCells numberBackhaus et al., 2001 [265]
norfloxacin69.624 hGrowth rate inhibitionCells numberBackhaus et al., 2001 [265]
ofloxacin82.824 hGrowth rate inhibitionCells numberBackhaus et al., 2001 [265]
Scenedesmus obliquusenrofloxacin45.1096 hGrowth rate inhibitionAbsorbanceQin et al., 2012 [266]
norfloxacin38.4996 hGrowth rate inhibitionCells numberNie et al., 2009 [267]
50.1896 hGrowth rate inhibitionCells numberLu et al., 2007 [268]
Scenedesmus dimorphusmoxifloxacin2696 hGrowth rate inhibitionAbsorbanceLi et al., 2023 [264]
Aquatic plants
Lemna minorciprofloxacin0.06257 dYield inhibitionDry weightEbert et al., 2011 [243]
0.2037 dGrowth rate inhibitionFronds numberRobinson et al., 2005 [245]
clinafloxacin0.0627 dGrowth rate inhibitionFronds numberRobinson et al., 2005 [245]
enrofloxacin0.1077 dYield inhibitionDry weightEbert et al., 2011 [243]
0.1147 dGrowth rate inhibitionFronds numberRobinson et al., 2005 [245]
flumequine2.4707 dGrowth rate inhibitionFronds numberRobinson et al., 2005 [245]
3.07 dYield inhibitionFronds numberZounková et al., 2011 [254]
levofloxacin0.0517 dGrowth rate inhibitionFronds numberRobinson et al., 2005 [245]
lomefloxacin0.1067 dGrowth rate inhibitionFronds numberRobinson et al., 2005 [245]
ofloxacin0.1267 dGrowth rate inhibitionFronds numberRobinson et al., 2005 [245]
Lemna gibbaciprofloxacin0.6987 dYield inhibitionWet weightBrain et al., 2004 [269]
levofloxacin0.5327 dYield inhibitionWet weightBrain et al., 2004 [269]
lomefloxacin0.1857 dYield inhibitionWet weightBrain et al., 2004 [269]
norfloxacin0.9137 dYield inhibitionFronds numberBrooks et al., 2008 [270]
ofloxacin0.5327 dYield inhibitionWet weightBrain et al., 2004 [269]
Crustaceans
Daphnia magnaciprofloxacin7.248 hImmobilizationImm. number Eluk et al., 2021 [271]
36.49348 hImmobilizationImm. number Dionísio et al., 2020 [272]
87.1448 hImmobilizationImm. number Dalla Bona et al., 2014 [273]
enrofloxacin7.948 hImmobilizationImm. number Eluk et al., 2021 [271]
16.7248 hImmobilizationImm. number Tolosi & De Liguoro 2021 [21]
3.1348 h + 10 dDelayed immobilizationImm. number Tolosi & De Liguoro 2021 [21]
16.3448 hImmobilizationImm. number Dalla Bona et al., 2014 [273]
flumequine7.1848 h + 10 dDelayed immobilizationImm. number Tolosi & De Liguoro 2021 [21]
gatifloxacin330.848 hImmobilizationImm. number Mala & Dutta 2019 [274]
gemifloxacin489.248 hImmobilizationImm. number Mala & Dutta 2019 [274]
levofloxacin2848 hImmobilizationImm. number Kergaravat et al., 2021 [275]
19.548 hImmobilizationImm. number Eluk et al., 2021 [271]
15.1148 hDelayed immobilizationImm. number Tolosi & De Liguoro 2021 [21]
lomefloxacin16648 hImmobilizationImm. number Luo et al., 2018 [276]
marbofloxacin5.448 hImmobilizationImm. number Eluk et al., 2021 [271]
moxifloxacin1448 hImmobilizationImm. number Kergaravat et al., 2021 [275]
norfloxacin8.748 hImmobilizationImm. number Eluk et al., 2021 [271]
ofloxacin31.7548 hImmobilizationImm. number Isidori et al., 2005 [259]
3648 hImmobilizationImm. number Eluk et al., 2021 [271]
Daphnia curvirostrisciprofloxacin4.4548 hImmobilizationImm. number Dalla Bona et al., 2014 [273]
enrofloxacin4.3348 hImmobilizationImm. number Dalla Bona et al., 2014 [273]
Ceriodaphnia dubiaciprofloxacin3648 hImmobilizationImm. number Kergaravat et al., 2021 [275]
enrofloxacin6048 hImmobilizationImm. number Kergaravat et al., 2021 [275]
levofloxacin3548 hImmobilizationImm. number Kergaravat et al., 2021 [275]
marbofloxacin3148 hImmobilizationImm. number Kergaravat et al., 2021 [275]
moxifloxacin2948 hImmobilizationImm. number Kergaravat et al., 2021 [275]
norfloxacin7348 hImmobilizationImm. number Kergaravat et al., 2021 [275]
ofloxacin17.417 dImmobilizationImm. number Isidori et al., 2005 [259]
Moina macrocopaciprofloxacin71.248 hImmobilizationImm. number Andrieu et al., 2015 [53]
enrofloxacin69.148 hImmobilizationImm. number Andrieu et al., 2015 [53]
Fish
Danio reriociprofloxacin62066 hEmbryonic mortalityDead numberHan et al., 2021 [277]
enrofloxacin15096 hEmbryonic mortalityDead numberÖzhan Turhan 2021 [278]
flumequine4048 hEmbryonic mortalityDead numberLancieri et al., 2002 [279]
gatifloxacin439366 hEmbryonic mortalityDead numberHan et al., 2021 [277]
levofloxacin543766 hEmbryonic mortalityDead numberHan et al., 2021 [277]
lomefloxacin143066 hEmbryonic mortalityDead numberHan et al., 2021 [277]
moxifloxacin60966 hEmbryonic mortalityDead numberHan et al., 2021 [277]
norfloxacin131166 hEmbryonic mortalityDead numberHan et al., 2021 [277]
Table 2. EC50 value of eleven fluoroquinolones expressed as Microcystis-equivalents and used for the construction of the normalized Species Sensitivity Distribution curve. Values in brackets represent the number of experimental data available in the literature. Cip: ciprofloxacin; Cli: clinafloxacin; Enr: enrofloxacin; Flu: flumequine; Gat: gatifloxacin; Lev: levofloxacin; Lom: lomefloxacin; Mox: moxifloxacin; Nor: norfloxacin; Ofl: ofloxacin; Sar: sarafloxacin; Mean: geometric mean. Data are obtained from [53,243,244,245,246,247,249,250,251,252,253,254,255,256,257,258,259,260,262,263,266,267,268,269,270,310].
Table 2. EC50 value of eleven fluoroquinolones expressed as Microcystis-equivalents and used for the construction of the normalized Species Sensitivity Distribution curve. Values in brackets represent the number of experimental data available in the literature. Cip: ciprofloxacin; Cli: clinafloxacin; Enr: enrofloxacin; Flu: flumequine; Gat: gatifloxacin; Lev: levofloxacin; Lom: lomefloxacin; Mox: moxifloxacin; Nor: norfloxacin; Ofl: ofloxacin; Sar: sarafloxacin; Mean: geometric mean. Data are obtained from [53,243,244,245,246,247,249,250,251,252,253,254,255,256,257,258,259,260,262,263,266,267,268,269,270,310].
CipCliEnrFluGatLevLomMoxNorOflSarMean
Chlorella sp.2600.58
(3)
2265.31
(1)
464.01
 
397.59
 
1021.03
Scenedesmus obliquus 920.41
(1)
976.62
 
948.10
Chlamydomonas
reinhardtii
3282.21
(1)
209
 
828.21
Scenedesmus
dimorphus
430.89
 
430.89
Scenedesmus
vacuolatus
6.63
(1)
311.83
 
1546.67
 
3942.86
 
335.09
Raphidocelis
subcapitata
744.7
(7)
68.57
(1)
63.27
(1)
10.92
(5)
372.49
(2)
122.04
 
513
 
206.01
 
1066.67
 
178.16
Desmodesmus
subspicatus
113.63
(1)
113.63
Anabaena
flos-aquae
1.08
(1)
3.53
(1)
600.00
(1)
124.44
 
23.10
Lemna gibba75.71
(1)
66.50
(1)
0.52
 
20.29
 
33.24
 
17.76
Lemna minor39.41
(3)
6.00
(1)
2.25
(2)
4.88
(2)
6.38
(1)
0.57
 
6
 
4.78
Microcystis
panniformis
1.47
(1)
1.47
Microcystis
aeruginosa
1
(2)
1
(1)
1
(1)
1
(2)
1
(1)
1
(1)
1
(1)
1
(1)
1
(1)
1
(1)
1
(1)
1.00
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pauletto, M.; De Liguoro, M. A Review on Fluoroquinolones’ Toxicity to Freshwater Organisms and a Risk Assessment. J. Xenobiot. 2024, 14, 717-752. https://doi.org/10.3390/jox14020042

AMA Style

Pauletto M, De Liguoro M. A Review on Fluoroquinolones’ Toxicity to Freshwater Organisms and a Risk Assessment. Journal of Xenobiotics. 2024; 14(2):717-752. https://doi.org/10.3390/jox14020042

Chicago/Turabian Style

Pauletto, Marianna, and Marco De Liguoro. 2024. "A Review on Fluoroquinolones’ Toxicity to Freshwater Organisms and a Risk Assessment" Journal of Xenobiotics 14, no. 2: 717-752. https://doi.org/10.3390/jox14020042

Article Metrics

Back to TopTop