Next Article in Journal
Fabrication of Superhydrophobic Porous Brass by Chemical Dealloying for Efficient Emulsion Separation
Previous Article in Journal
Research on the Effects of Drying Temperature for the Detection of Soil Nitrogen by Near-Infrared Spectroscopy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Quantification of Metal(loid)s in Lubricating Eye Drops Used in the Treatment of Dry Eye Disease

by
Marcelo de Oliveira
1,2,
Elaine S. de Pádua Melo
2,
Thaís Carvalho da Silva
2,
Carla Maiara Lopes Cardozo
1,2,
Igor Valadares Siqueira
2,
Mariana Pereira Hamaji
2,
Vanessa Torres Braga
1,
Luiz Fernando Taranta Martin
2,
Alessandro Fonseca
2 and
Valter Aragão do Nascimento
1,2,*
1
Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil
2
Group of Spectroscopy and Bioinformatics Applied to Biodiversity and Health, Postgraduation Program in Health and Development in the Midwest Region, School of Medicine, Faculty of Medicine, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(18), 6508; https://doi.org/10.3390/molecules28186508
Submission received: 17 August 2023 / Revised: 1 September 2023 / Accepted: 3 September 2023 / Published: 7 September 2023

Abstract

:
The aim of the study was to evaluate the presence of metal(loid)s in lubricating eye drops used in the treatment of dry eye disease. The concentrations of Al, As, Ba, Cd, Co, Cu, Cr, Pb, Fe, Mg, Mn, Mo, Ni, Se, V, and Zn were determined in 19 eye drop samples using inductively coupled plasma optical emission spectrometry (ICP OES). The limit of detection (LOD) and limit of quantification (LOQ) values for the quantified elements ranged from 0.0002–0.0363 (mg/L) and 0.0007–0.1211 (mg/L), respectively. High values of concentrations of Al (2.382 µg/g), As (0.204 µg/g), Ba (0.056 µg/g), Cd (0.051 µg/g), Co (1.085 µg/g), Cr (0.020 µg/g), Cu (0.023 µg/g), Fe (0.453 µg/g), Mg (24.284 µg/g), Mn (0.014 µg/g), Mo (0.046 µg/g), Ni (0.071 µg/g), Pb (0.049 µg/g), Se (0.365 µg/g), V (0.083 µg/g), and Zn (0.552 µg/g) were quantified in samples of eye drops with and without preservatives. The concentrations of As (5 samples) and Cd (3 samples) were higher than those allowed by the Brazilian Pharmacopoeia for impurities (parenteral use). The value of Co content (µg/g) in a sample was higher than the value established by the International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use (ICH Q3D (R2)) in the parenteral route. The daily eye drop instillation exposure (µg/day) was below the values from the parenteral-permitted daily exposure (PDE) set by the ICH Q3D guideline (R2). The presence of heavy metals in eye drops is an alert to regulatory agencies in several countries so that control and inspections can be carried out.

1. Introduction

According to the World Health Organization (WHO, 2019), eye conditions that do not typically cause vision impairment, such as dry eye (DE), must not be overlooked [1]. Therefore, several countries have sought solutions to prevent and treat this disease [2]. Dry eye disease, also called dry eye syndrome, dysfunctional tear syndrome, and keratitis sicca, is a multifactorial disease of the ocular surface characterized by the loss of tear film homeostasis [3,4], which affects millions of people [4], with adults over 40 years of age and women being the most affected [5]. The most common causes of dry eye disease are age, menopause, hormonal changes, autoimmune diseases, inflamed eyelid glands, and allergic eye diseases. Further causes include the constant or excessive use of computers, exposure to air conditioning, the use of contact lenses, and the ingestion of drugs, such as benzodiazepines, antidepressants, antihistamines, and analgesics [3].
The diagnosis of DE is made from the identification of the symptoms and clinical tests. However, this disease has multiple symptoms and signs that often do not correlate with one another. As a result, greater attention is being paid to diagnosing and imaging technologies to assess the type and severity of dry eye disease [5,6,7]. Conventional treatments incorporate lubricating eye drops or artificial tears, which can be effective in mild and moderate cases [8]. In addition, lubricating eye drops have different levels of osmolarity, pH, and viscosity; however, they contain the same aqueous component that is important in lubricating the ocular surface [9].
The eye drops used in dry eye disease may contain preservatives in order to avoid contamination of bacteria in the vials after opening [10]. Although, chronic use of preservatives can cause significant damage to the eye tissues [11]. On the other hand, lubricating eye drops without preservatives can minimize the effects of irritation and ocular toxicity; however, they are expensive and the bottle is difficult to handle, especially for elderly patients [12].
Previous studies have reported that metals and metalloids play an important role in the pathogenesis of several ophthalmological disorders, such as Cd, Cu, Fe, Hg, Ni, and Zn in glaucoma [13,14], as well as the elements As, Cd, Co, Cr, Cu, Fe, Hg, Ni, and Pb in age-related macular degeneration (AMD) [15,16,17], Al, Bi, Cd, Cs, Fe, Pb, Te, and Tl in cataracts [18,19], and Cd, Hg, and Pb in dry eye disease [20,21,22,23,24]. Furthermore, there is a significant level of heavy metals in the aqueous humor [14,25], as well as in the lens [19,25,26], and in the pigment epithelium of the retina and choroid [17,27]. However, clinical studies on the toxicity of heavy metals and metalloids in eye drops with preservatives and without preservatives are scarce.
The impurities present in eye drops can be organic, inorganic, and residual solvents. Most of these impurities occur due to manufacturing processes, degradation, storage conditions, excipients, or contamination [28,29,30]. In this context, some countries have pharmacopoeias that establish quality parameters and analysis methods for supplies and medicines [31,32,33]. However, in the absence of information in their pharmacopoeias, some countries resort to the guidelines for elemental impurities (ICH Q3D), which are established by the International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use (ICH) [34,35,36].
Since metals have various oxidation states, their toxicities are different [37]. Cr (III) is essential to health human, while Cr (VI) is highly toxic [38]. In addition, pentavalent methylated arsenicals are less toxic, while trivalent methylated species are highly cytotoxic [38]. Various transition metal ions are toxic and produce various types of diseases in the body. Metal ions are present in water, soil, sediment, and blood samples and their valence state plays a crucial role for absorption and dynamics behavior in the biological context [37,39]. In light of the above, it has been observed that more research is needed to investigate the values of the concentrations of metal(loid)s in various types of medication and compare them with the values established by the regulatory agencies.
The ICH Q3D guideline, currently in version 2 (R2), presents a process to assess and control elemental impurities in the drug product administered orally, parenterally, and inhaled; that is, it establishes the limits of permitted daily exposure (PDE) in µg/day for each element of toxicological concern and the concentration-permitted levels of impurities in drugs, drug substances, and excipients [40,41,42]. However, to date, permitted concentrations and daily exposure of metal(loid)s for the ophthalmic route have not been established [31,40,43].
Motivated by the results published in Refs. [13,14,15,16,17,18,19,20,21,22,23,24], who demonstrated that potential risks to ocular health may be related to the presence of heavy metals, our study aimed to evaluate, for the first time, the presence of Al (aluminum), As (arsenium), Ba (barium), Cd (cadmium), Co (cobalt), Cr (chromium), Cu (copper), Fe (iron), Mg (magnesium), Mn (manganese), Mo (molybdenum), Ni (nickel), Pb (lead), Se (selenium), V (vanadium), and Zn (zinc) in Brazilian lubricating eye drops with and without preservatives used in the treatment of dry eye disease. In addition, the contents of chemical elements in the studied eye drop samples were compared with the values of allowed concentrations of permitted concentration of elemental impurities established by the Brazilian Pharmacopoeia (BP) [31], as well as elemental impurity values set by the ICH Q3D (R2) [40], and PDE values recommended by the ICH Q3D guideline (R2) in the parenteral route [40].

2. Results

The results were presented as follows: Section 2.1 outlines the results of the analyte addition and recovery test; the result of the calibration curve is shown in Section 2.2; in Section 2.3, data on the concentrations of heavy metals and metalloids quantified (µg/g) in samples of lubricating eye drops and the comparison with the values of permissible concentrations of elemental impurities established in the parenteral route of Brazilian Pharmacopeia (BP) [31] and ICH guideline Q3D (R2) [40] are presented. The latter data (Section 2.4) provides data on the exposure to heavy metals and metalloids during the daily instillation of lubricating eye drops in the eyes (µg/day) and the comparison with the PDE values established by the ICH Q3D guideline (R2) via the parenteral route.

2.1. Analyte Addition and Recovery Test

A spike was generated by adding a known amount (0.5 mg/L) of analyte to a sample, testing the spiked sample. The recovery results were as follows: Al 91.37%, As 90.86%, Ba 95.38%, Cd 96.12%, Co 90.57%, Cr 99.05%, Cu 91.69%, Fe 96.19%, Mg 97.13%, Mn 95.31%, Mo 90.23%, Ni 92.70%, Pb 95.85%, Se 97.10%, V 91,12%, and Zn 98.17%.

2.2. Calibration Curves

The range values of the detection limits (LOD), quantification limits (LOQ), and correlation coefficients (R2) for the detected elements were 0.0002–0.0363 (mg/L), 0.0007–0.1211 (mg/L), and 0.9968–0.9996, respectively. Table 1 presents the parameters of the calibration curve and the values obtained for the LOD, LOQ, and R2.

2.3. Concentration of Metal(loid)s in Lubricating Eye Drops

In this study, the concentrations of metal(loid)s in 19 commercial samples of lubricating eye drops used in the treatment of dry eye disease in Brazil were analyzed using optical emission spectrometry with inductively coupled plasma (ICP OES). A total of fourteen samples with preservatives (samples 1–14) and five without preservatives (samples 15–19) were analyzed. Table 2 shows the concentrations of Al, As, Ba, Cd, Co, Cr, Cu, Fe, Mg, Mn, Mo, Ni, Pb, Se, V, and Zn quantified in lubricating eye drops with preservatives and without preservatives compared with the permitted concentrations of elemental impurities established by the BP [31] and ICH Q3D (R2) guideline for parenteral administration [40]. Figure 1 shows the values of metal(loid) concentrations quantified in the eye drop samples with and without preservatives. The concentration of Al in the eye drop samples 2–11, 16, 17, and 19, Cr in samples 1–8, 10, and 12–18, Fe in samples 2, 4, 5, 7, 9–11, 14, 16, and 17, Mg in samples 4, 6, 17, 18, and 19, and Pb in samples 1, 3–7, and 14–19 are all below the limit of detection (<LOD).

2.4. Exposure to Metal(loid)s by Daily Instillation of Lubricating Eye Drops

Exposure to metal(loid)s through the daily instillation of lubricating eye drops was obtained by multiplying the concentration of quantified elements in the eye drops (Table 2) by the maximum daily dose of the sample [44]. In addition, the results obtained regarding the exposure to metal(loid)s through the daily instillation of lubricating eye drops were compared with the PDE for elemental impurities of the ICH Q3D (R2) guideline for the parenteral route (Table 3). Figure 2 shows the values obtained through the daily instillation of lubricating eye drops in the eyes (µg/day) with and without preservatives.

3. Discussion

Spike tests generated information about the recovery of the spiked samples. Since there are no certified reference metal and metalloids samples in eye drops, spike recovery tests must be used for quality control. The values of LOD and LOQ are displayed in Table 1. As can be seen, the LODs and LOQs allowed for the determination of both metals and metalloids at the required levels based on the IUPAC [45].
The concentrations of Al, As, Ba, Cd, Co, Cr, Cu, Fe, Mg, Mn, Mo, Ni, Pb, Se, V, and Zn in lubricating eye drops from different manufacturers (Table 2) decreased in values as follows:
  • eye drops 1 (with preservatives): Mg > Zn > Se > Fe > As > Al > Cd > Ni > Mo > Ba > Co > Cu > V > Mn;
  • eye drops 2 (with preservatives): Se > Mg > As > Zn > Ni > Cd > Mo > Pb = Co > Ba > V > Mn > Cu;
  • eye drops 3 (with preservatives): Se > Mg > Fe > As > Z > Cd > Ba > Mo > Ni > Co > Cu > V > Mn; eye drops 4 (with preservatives): Se > As > Zn > Cd > Mo > Ni > Ba > Co = V > Cu > Mn;
  • eye drops 5 (with preservatives): Mg > Co > Se > As > Cd > Zn > V > Mo > Ni > Ba > Mn > Cu;
  • eye drops 6 (with preservatives): Se > As > Zn > Cd > Mo > Ni > Co > Fe > Ba > V = Cu > Mn;
  • eye drops 7 (with preservatives): Mg > Se > As > Zn > Ni = Cd >Mo > V > Co > Ba > Mn > Cu;
  • eye drops 8 (with preservatives): Mg > Se > Zn > As > Ni > Cd > Mo > Co > Ba > Pb > V > Cu > Mn > Fe;
  • eye drops 9 (with preservatives): Mg > Se > As > V > Ni > Zn > Cd > Pb > Mo > Co > Ba > Cu > Mn > Cr;
  • eye drops 10 (with preservatives): Mg > Se > Zn > As > Ni > Cd > Mo > Pb > Co > Ba > V > Mn > Cu;
  • eye drops 11 (with preservatives): Mg > Zn > Se > As > Ni > Cd > Mo > Co > Ba > V > Pb > Mn > Cr > Cu;
  • eye drops 12 (with preservatives): Mg > Al > Fe > Se > As > Ni > V > Zn = Cd > Mo > Co = Ba > Pb > Mn > Cu;
  • eye drops 13 (with preservatives): Al > Fe > Se > Mg > Zn > As > Ni > Cd > V > Mo > Ba > Co > Pb > Mn > Cu;
  • eye drops 14 (with preservatives): Se > Mg > As > Al > Cd > Ni = Zn > Mo > Ba > Co > Mn > V > Cu;
  • eye drops 15 (without preservatives): Se > Al > Fe > As > Cd > Co > Ni = Mo > Ba = Mg > Zn > V > Cu = Mn;
  • eye drops 16 (without preservatives): Se > As > Cd >Ni > Ba > Mo > Zn > Co > V > Mn > Cu;
  • eye drops 17 (without preservatives): Se > As > Ba > Cd > Mo = Ni > Zn > Cu > Co > V > Mn;
  • eye drops 18 (without preservatives): Al > Fe > Se > Mg > Cd > Zn > As > Ni > Co > V > Mo > Ba > Cu = Mn;
  • and eye drops 19 (without preservatives): Se > Zn > As > Fe > Cd > Co > Ba = Ni > Mo > Cr > V > Mn > Cu.
Values of the concentrations of metal(loid)s quantified in lubricating eye drops with preservatives and without preservatives are shown in Figure 1. Higher concentrations were obtained for several elements, such as Mg, Al, and Co. The statistical results for all data in Table 2 are shown in Supplementary Materials Table S1. According to the ANOVA, there is a statistically significant difference in the concentration means of Al, As, Ba, Cd, Co, Cr, Cu, Fe, Mg, Mn, Mo, Ni, Pb, Se, V, and Zn in all eye drops with preservatives and without preservatives. Tukey’s test identifies which means are significantly different from the others. In Table S1 (Supplementary Materials), when the letters are the same, there are no statistical differences; on the other hand, when the letters are different, there is a statistical difference between the values.
Figure 2 shows the values of exposure to metal(loid)s through the daily instillation of lubricating eye drops with and without preservatives. Elevated exposure values were deemed to be due to higher concentrations of the elements Mg, Al, and Co.
Table S2 (Supplementary Materials) shows the statistical test used in view of the results of Table 3. According to the ANOVA statistical test and Tukey’s test, there are significant differences in the average values of daily exposure to metal(loid)s between the samples of lubricating eye drops with preservatives and the samples without preservatives (p < 0.05) (Table S2 in the Supplementary Materials). For all values of daily instillation with the same letter, the difference between the means is not statistically significant.
According to Table 2, the content of Al ranged from 0.073 µg/g to 2.382 µg/g. There were levels of Al detected in eye drop samples 1, 12, 13, and 14 with preservatives, and in eye drops 15 and 18 without preservatives. As shown in Table 2, there were no values of Al detected in the parenteral drugs established by the BP and ICH Q3D guideline (R2). On the other hand, exposure to Al through the daily instillation of lubricating eye drops in Table 3 ranged from 0.037 µg/day (eye drops 14 with preservatives) to 0.793 µg/day (eye drops 18 without preservatives). There were no PDE values for the elemental impurities established by the ICH Q3D (R2) parenteral guideline. Nevertheless, Al can potentiate oxidative, inflammatory events and cause eventual tissue damage [46]. According to research involving animals, intraperitoneal application of aluminum chloride causes retinal toxicity and loss of the outer segments of photoreceptors [47].
The concentration of As in the eye drop samples ranged from 0.036 µg/g to 0.204 µg/g (Table 2). In eye drops 12 (0.147 µg/g) and 13 (0.147 µg/g), both with preservatives, the concentration values for As were close to the permitted concentrations of elemental impurities established by the BP for the parenteral route (0.15 µg/g), while the contents in eye drops 2 (0.169 µg/g), 8 (0.204 µg/g), 9 (0.195 µg/g), and 10 (0.184 µg/g) with preservatives and eye drops 16 without preservatives (0.178 µg/g) were all above the concentration value defined in the BP for parenteral use (See Table 2). In addition, all eye drops had concentrations of As below the limit established by the ICH Q3D guideline (R2) for this element in the parenteral route (1.5 µg/g). According to Table 3, the values of exposure to As due to the daily instillation of eye drops ranged from 0.012 µg/day (eye drops 18 without preservatives) to 0.109 µg/day (eye drops 10 without preservatives). The values of exposure to As in all eye drop samples were below the PDE limits established by the ICH Q3D guideline (R2) (15 µg/day). However, although the value of daily exposure to As through the instillation of lubricating eye drops is below the value established by the ICH Q3D guideline (R2), chronic exposure to this element can cause dermal effects, cardiovascular diseases, diabetes, cancers [48], eye irritation [49], conjunctivitis [50], and visual loss [51].
In Table 2, the content of Ba in the eye drop samples ranged from 0.021 µg/g to 0.056 µg/g. There are no Ba values established by the BP for impurities in parenteral drug administration. However, all eye drop samples showed concentrations of Ba below the values allowed by the ICH Q3D guideline (R2) via the parenteral route (70 µg/g). According to Table 3, the exposure to Ba caused by the daily instillation of lubricating eye drops ranged from 0.006 µg/day (eye drops 19 without preservatives) to 0.015 µg/day (eye drop samples 10 and 12 with preservatives); moreover, these values are below the PDE (700 µg/day) established by the ICH Q3D guideline (R2) in the parenteral route. The exposure to barium compounds can cause hypokalemia, cardiovascular diseases, muscle weakness and paralysis [52,53], and eye pain [54], as well as a toxic ocular inflammatory reaction (toxic anterior segment syndrome—TASS) [55]. In rabbits, topical administration of Ba resulted in mild skin irritation and severe eye irritation [56].
The concentration of Cd in the eye drop samples ranged from 0.044 µg/g to 0.051 µg (Table 2). The Cd contents in eye drops 1 (0.049 µg/g), 6 (0.048 µg/g), 7 (0.048 µg/g), 8 (0.049 µg/g), 10 (0.049 µg/g), and 12 (0.049 µg/g) with preservatives, and eye drops 15 (0.049 µg/g) and 16 (0.048 µg/g) without preservatives were close to the concentration values of impurities in drugs allowed by the parenteral BP (0.05 µg/g), while the concentrations in eye drops 2, 3, and 9 (0.051 µg/g) with preservatives are above the allowed concentration of impurities established by the BP for parenteral use. All samples showed cadmium concentrations below the concentration value allowed by the ICH Q3D guideline (R2) parenterally (0.2 µg/g) (Table 2). Regarding Cd exposure, daily instillation values ranged from 0.009 µg/day (eye drops 16,17, and 19 without preservatives 16) to 0.030 µg/day (eye drops 12 with preservatives); all daily instillation values were below the PDE defined by the ICH Q3D guideline (R2) (2 µg/day) (Table 3). In the body, Cd can cause health risks, such as acute and chronic intoxication, pathological changes in the organs, and cardiovascular, kidney, bone, liver disease, cancer [57], and eye problems [13,16,17,18], such as cataracts [26] and glaucoma, especially in men [13]. Park et al. [15] suggested that Cd, Pb, and Hg may negatively influence AMD. On the other hand, the study by Liou et al. [20], involving 59 welders and 25 administrative staff from a shipyard in northern Taiwan, concluded that Cd found in urine and Pb in welders’ toenails (exposed group) may be related to dry eye disease. In fact, according to a study carried out by Choi et al. [23], considering the exposure of women to phthalates and heavy metals and diagnosed and non-diagnosed with dry eye disease, women diagnosed with dry eye disease have higher concentrations of Cd in their urine and blood compared to those not diagnosed.
As shown in Table 2, the content of Co in the eye drops ranged from 0.013 µg/g to 1.085 µg/g. In addition, there are no limits for Co concentrations established by the BF. Only eye drops 5, containing preservatives, had a concentration value greater than that established by the ICH Q3D guideline (R2) parenterally (0.5 µg/g). In Table 3, the values of exposure to Co considering the daily instillation of eye drops ranged from 0.003 µg/day (eye drops 17 without preservatives) to 0.554 µg/day (eye drops 5 with preservatives). The PDE values for elemental impurities established by the ICH Q3D guideline (R2) in the parenteral route are 5 µg/day. After comparison, it was observed that the daily instillation values considering Co in Table 3 were below those established by the ICH Q3D guideline (R2). However, excessive levels of Co in the body can cause cancer [58], peripheral neuropathy, hearing loss, cognitive deficits, cardiomyopathy, hypothyroidism, and visual changes [59,60]. Junemann et al. [16] quantified the levels of selected metal ions in the aqueous humor of eyes affected by AMD using flow injection inductively coupled plasma mass spectrometry (FI-ICP-MS) and observed that patients with AMD had significant levels of Cd, Co, Fe, and Zn, in addition to reduced levels of Cu, when compared to patients without AMD. One study reported that a 39-year-old woman presented with blurred vision in the left eye and bilateral ocular discomfort following the bilateral implantation of hip prostheses that had Co in their constitution [61]. In 2020, Garcia and collaborators published a case report of a 59-year-old woman who presented symptoms, such as blurred vision, white spots in both eyes, hypothyroidism, cardiomyopathy, and neuropathy, after reattachment of a hip prosthesis implant with a Co piece [59]. In both cited studies, the hypothesis was that implant-related Co toxicity occurred. Finally, Lim et al. [62] reported that elevated serum Co levels may be related to reversible and irreversible damage that leads to visual loss, such as neuropathy and atrophy of the optic nerve and abnormal electrophysiological functioning of the retinal tissue and the retinal pigment epithelium, in addition to abnormal choroidal perfusion.
As shown in Table 2, the content of Cr in eye drops 1–8, 10, 12–18, and 19 are below the detection limit. However, the Cr content in eye drops 9 (0.004 µg/g) and 11 (0.004 µg/g) with preservatives and eye drops 19 (0.020 µg/g) without preservatives were all below the permissible BP concentration (2.5 µg/g) and ICH guideline Q3D guideline (R2) through the parenteral route (110 µg/g). According to the results shown in Table 3, exposure to Cr ranged from 0.002 µg/day (eye drops 9 and 11 with preservatives) to 0.004 µg/day (eye drops 19 without preservatives). In Table 3, the exposure values for the instillation of eye drops considering Cr were all below the PDE for elemental impurities defined by the ICH Q3D guideline (R2) parenterally (1100 µg/day). Although these concentrations are low when compared to the limits established by the BP and ICH Q3D guideline (R2), exposure to Cr can cause hepatotoxicity, nephrotoxicity, dermatotoxicity, carcinogenicity [63], and inflammation of the digestive tract followed by necrosis, dermatitis, chronic painless ulcers, teeth and tongue am-amery, irritation of the mucous membranes, respiratory allergies, and cancer at the level of the respiratory system [64]. Exposure to hexavalent Cr can cause eye irritation, corrosion, and ulceration. [65,66].
The contents of Cu in the eye drops ranged from 0.002 µg/g (eye drops 14 with preservative) to 0.023 µg/g (eye drops 17 without preservative), being below the concentration allowed by the BP (25 µg/g) and ICH Q3D guideline (R2) parenterally (30 µg/g) (Table 2). Exposure to Cu due to the daily instillation of eye drops, whose values ranged from 0.001 µg/day (eye drops 11 and 14 with preservatives, and 16 and 19 without preservatives) to 0.007 µg/day (eye drops 1, 8 and 9 with preservatives), were below the PDE for elemental impurities defined by the ICH Q3D guideline (R2) in the parenteral route (300 µg/day) (Table 3). Cu plays a key role in the biochemistry of the human nervous system [67], being important for healing and slowing cataract growth [68]. On the other hand, intraocular foreign bodies containing Cu can cause inflammation, damage to the cell membranes and mitochondria in the retina [14], aseptic abscesses, cataracts, vitreous liquefaction and retraction, retinal damage and detachment, ocular hypotension [69], and corneal opacities [70].
In Table 2, the contents of Fe ranged from 0.013 µg/g (eye drops 8 with preservatives) to 0.453 µg/g (eye drops 18 without preservatives). However, there are no Fe concentration values in drugs allowed by the BP and ICH Q3D guideline (R2) parenterally. As shown in Table 3, exposure to Fe due to the daily instillation of eye drops ranged from 0.006 µg/day to 0.256 µg/day (drops 8 and 13 with preservatives). However, there are no Fe values established by the ICH Q3D (R2) parenteral guideline for parenteral Fe. This element is involved in several biological processes, but the cerebral accumulation or the decrease in intracellular Fe can impair the functioning of several functions in the central nervous system (CNS) and cause cell death [71]. Fe affects the retina [72] and its overload augments stage I and stage II of tumor promotion in murine skin [73]. Chronic accumulation of Fe in the retina can cause AMD and also influence other ocular conditions, such as hereditary aceruloplasminemia, pantothenate kinase-associated neurodegeneration, intraocular hemorrhage [74], and glaucoma [14].
The concentration of Mg ranged from 0.021 µg/g (eye drops 15 without preservatives) to 24.284 µg/g (eye drops 9 with preservatives). However, there are no Mg concentration values in drugs allowed by the BP and ICH Q3D guideline (R2) parenterally. According to Table 3, exposure to Mg due to the daily instillation of eye drops ranged from 0.010 µg/day (eye drops 15 without preservatives) to 11,025 µg/day (eye drops 9 with preservatives). There are no PDE values for Mg as elemental impurities established by the ICH Q3D (R2) guideline for the parenteral route. In addition, the exposure of eye drops 9 with preservatives was higher in relation to the other heavy metals and metalloids analyzed. In this study, the highest values found for concentration and exposure through the daily instillation of eye drops were in relation to Mg. In the human body, Mg is important for maintaining the structural and functional integrity of the lens, in addition to playing a significant role as a cofactor for enzymes involved in the production and hydrolysis of adenosine triphosphate (ATP). Mg deficiency leads to the accumulation of Ca in the lens, which can cause opacification, along with the release of nitric oxide that produces nitrogen free radicals that are capable of causing oxidative damage [25]. In toxic situations, the most common symptoms are diarrhea, nausea and vomiting, muscle weakness, and low blood pressure, but as levels increase there is a loss of deep tendon reflexes, blockages of the sinus or atrioventricular node, respiratory paralysis, and, eventually, cardiac arrest [75].
Mn levels in eye drops 5 (0.006 µg/g), 8, and 9 (0.014 µg/g) with preservatives are below the concentration value established by the BP for impurities in parenteral drugs (25 µg/g). However, there are no values for Mn established by the ICH Q3D (R2) guideline for parenteral administration. Table 3 shows that exposure to Mn due to the daily instillation of eye drops ranged from 0.001 µg/day in samples 16 and 17 without preservatives; on the other hand, in eye drops 8 and 13 with preservatives it was 0.007 µg/day. There are no PDE values for elemental impurities set out in the ICH Q3D (R2) parenteral guideline. However, in the human body, Mn is important for healing and in delaying cataract growth [68]; meanwhile under high concentrations, Mn can cause toxicity to the reproductive, cardiac, respiratory, and central nervous systems [64]. According to animal studies, there are toxic effects of manganese ion eye drops to the ocular anterior segment [76]. Khosla et al. [77] studied the effect of Mn on the retina of rabbits and found that it is potentially retinotoxic, causing a selective effect on photoreceptors and ganglion cells. In addition, studies involving humans proved that patients with age-related ocular disorders have higher Mn concentrations when compared to a control group [78].
The Mo content in eye drops ranged from 0.025 µg/g (eye drops 18 without preservatives) to 0.046 µg/g (eye drops 9 with preservatives); however, such values were below the permissible BP concentration (2.5 µg/g) and ICH Q3D guideline (R2) for the parenteral route (150 µg/g). Exposure to Mo considering the daily instillation of eye drops ranged from 0.005 µg/day (eye drops 17 without preservatives) to 0.026 µg/day (eye drops 10 with preservatives). Therefore, Mo exposure values (Table 3) were found to be below those of the PDE established by the ICH Q3D guideline (R2) for the parenteral route (1500 µg/day). However, in studies involving workers exposed to high levels of Mo, it was found that the inhalation of dust containing this metal can cause lung problems [79]. According to Khosla et al. [77], retinotoxic effects of molybdenum on rabbit retina were observed (in an experimental study). Results involving humans corroborate those published with animal models [77]. In fact, Ceylan et al. [78] found an elevated Mo level in patients with pseudoexfoliation syndrome. In another study, molybdenum trioxide, a soluble molybdenum compound, was found to have an irritating effect on the skin, as well as the mucous and eyes [80].
The concentration of Ni ranged from 0.023 µg/g to 0.071 µg/g, but these values were found to be below the concentration value of impurities in drugs established by the BP (2.5 µg/g) and ICH Q3D guideline (R2) for parenteral administration (2 µg/g) (Table 2). Considering the daily instillation of eye drops (Table 3), the results indicate that exposure to Ni ranged from 0.005 µg/day (eye drops 17 without preservatives) to 0.040 µg/day (eye drops 10 with preservatives). However, these daily eye drop instillation values were found to be below the PDE for Ni (20 µg/day) established by the ICH Q3D guideline (R2) for the parenteral route. Ni is essential for the human body and is part of enzymatic and hormonal activity, and aids in promoting the structural stability of biological macromolecules and metabolism. Its toxicity causes headaches, nausea, vomiting, apathy, diarrhea, fever, skin disorders, carcinomas in the nasal cavities and lower respiratory tract [64], and conjunctivitis [81]. According to an animal study, cellular anomalies in the retina of fish eyes exposed to Ni affect the primary functions of the retina and lead to visual loss or low vision [82]. A study with eyeballs from human cadavers found that the levels of Ni, As, Cd, Cr, and Pb were elevated in late AMD when compared to the control groups [17].
In Table 2, the concentration of Pb in the eye drop samples ranged from 0.009 µg/g (eye drops 11 with preservatives) to 0.049 µg/g (eye drops 9 with preservatives). However, this concentration was below the allowed concentration for impurities in drugs established by the BP (0.1 µg/g) and ICH Q3D guideline (R2) for the parenteral route (0.5 µg/g). According to these results, considering the daily instillation of eye drops (Table 3), the exposure to Pb that varied from 0.004 µg/day to 0.022 µg/day in eye drops 2, 8–12, and 13, all with preservatives, were below the PDE for Pb (5 µg/day) established by the ICH Q3D guideline (R2) in the parenteral route. However, the accumulation of Pb in the body can cause health risks and be associated with eye diseases [14,27,83], such as cataracts [18], AMD [15,17], and dry eye disease [20,24].
As shown in Table 2, the contents of Se ranged from 0.211 µg/g to 0.365 µg/g. There are no Se values for impurities in drugs established by the BP for the parenteral route. In addition, all eye drop samples had concentrations below the concentration allowed in the parenteral route (8 µg/g) established by the ICQ Q3D guideline (R2). Daily exposure to Se ranged from 0.055 µg/day (eye drops 17 without preservatives) to 0.188 µg/day (eye drops 12 with preservatives) (Table 3). Furthermore, the Se exposure values in Table 3 were all below the PDE for Se (80 µg/day) defined by the ICH guideline Q3D (R2) for the parenteral route. However, exposure to Se at high levels can cause selenosis, respiratory tract irritation, bronchitis, difficulty breathing, stomach pains, and coughing [84]. In studies with mice, it was observed that after topical application of selenium sulfide erythema, skin irritation, acanthosis, and severe skin damage occurred [85]. According to the results of the study by Bruhn et al. [86], there is a relationship between glaucoma and selenium levels in plasma and the aqueous humor. In another experimental study involving animals, it was verified that any amount of Se above the concentrations necessary for the synthesis of selenoproteins is toxic and can cause cataracts [87].
The concentration values of V in the eye drops ranged from 0.008 µg/g (eye drops 14 with preservatives) to 0.083 µg/g (eye drops 9 with preservatives) (Table 2). In addition, this concentration was below that allowed by the BP for impurities in drugs (2.5 µg/g) and by the ICH Q3D guideline (R2) for the parenteral route (1 µg/g). Table 3 shows that exposure to V due to daily instillation ranged from 0.002 µg/day (eye drops 16, 17, and 19 without preservatives) to 0.038 µg/day (eye drops 9 with preservatives). Furthermore, these values were found to be lower than those established by the ICH Q3D (R2) guideline for Se (10 µg/day). In situations of V poisoning, people may have symptoms, such as headaches, hand tremors, high blood pressure, green tongue, cough, palpitations, wheezing, ear and throat irritation [88], skin rash [89], eye irritation, and conjunctivitis [90]. Furthermore, studies have shown that inhalation of V produces damage to the retina detected through the damage markers GFAP (glial fibrillary acidic protein) and GS (glutamine synthetase) and might interfere with vision [91,92].
The concentration of Zn ranged from 0.019 µg/g (eye drops 15 without preservatives) to 0.552 µg/g (eye drops 11 with preservatives) (Table 2). However, there are no established concentration values for Zn in impurities in drugs established by the BP and ICH Q3D (R2) guideline for the parenteral route. Exposure to Zn ranged from 0.004 µg/day (eye drops 16 without preservatives) to 0.265 µg/day (eye drops 11 with preservatives) (Table 3). There are no PDE values for Zn established by the ICH Q3D guideline (R2) considering the parenteral route. Zn is essential for the immune system [64] and acts to prevent the progression of AMD in advanced stages [15,93], in addition to influencing muscle function and vision [83]. However, its excess in the lens may be related to the presence of high molecular weight proteins that are considered precursors of insoluble protein aggregates that cause lens opacification [25]. Ocular exposure to zinc salts can cause irritation, pain, corneal ulcerations, edema and burns, hyperemia, hemorrhage, bullous keratopathy, glaucoma, cataract formation, discrete gray spots on the lens, tearing, significant reduction in acuity visual with hemorrhage, and conjunctival inflammation [94].
As we observed in the results and discussion above, although the concentrations of several elements are below the limit values that have been established by regulatory agencies, such as the BP and ICH Q3D (R2), it does not mean that their use is safe. Metals and metalloids can be dangerous for human health as they have a long biological half-life and are not biodegradable, and some are toxic even at very low concentrations [95,96,97].
This study shows that there is a presence of toxic metals in eye drops used in the treatment of dry eye; therefore, regulatory agencies must establish a PDE level in micrograms per day (µg/day) through the instillation of lubricating eye drops in the eyes.

4. Materials and Methods

4.1. Sample Collection

In this study, a total of 95 eye drop samples from different manufacturers were purchased in December 2021 from pharmacies in the city of Campo Grande, state of Mato Grosso do Sul, Brazil. Only samples of lubricating eye drops with a volume of 10 mL and available in drops were selected, with 5 samples of manufacturers without preservatives (25 samples from each batch and same eye drop manufacturer) and 14 samples of manufacturers with preservatives (70 samples from each eye drop manufacturer). From each company, 5 copies of the same batch were purchased; however, samples from each batch and from the same company were mixed to obtain a representative sample for analysis.

4.2. Preparation for the Analysis

All materials used in the study, such as Falcon glassware and plastic tubes, underwent a chemical demineralization process before being used. The materials were placed in a solution of Extran (5%, v/v) and nitric acid (10% concentration, Merck, Darmstadt, Germany) for a minimum period of 24 h, following which they were rinsed in ultrapure water and dried in an oven at 42 °C.

4.3. Acid Digestion of Samples

Approximately 0.5 mL of each sample was placed in a glass test tube, followed with the additions of 2.0 mL of HNO3 (65%, Merck, Darmstadt, Germany), and 1.0 mL of H2O2 (35%, Merck, Darmstadt, Germany).
The samples were digested in an open digestion system for 40 min at a temperature of 80 °C. Subsequently, all samples were transferred from glass test tubes to Falcon tubes, where 2.5 mL of ultrapure water (conductivity 18.2 MΩcm (Millipore), Biocel, Germany) was added.
All steps of the digestion analysis were performed in triplicate and the analytical blanks were prepared following the same procedure used in the samples.

4.4. Elementary Analysis Using ICP OES

The contents of 16 elements (Al, As, Ba, Cd, Co, Cu, Cr, Pb, Fe, Mg, Mn, Mo, Ni, Se, V, and Zn) were quantified using ICP OES (Thermo Fisher Scientific, Bremen, Germany, model iCAP 6300 Duo). Instrumental and operational parameters for ICP OES are shown in Table 4.
An analyte addition and recovery test (spike test) was performed, in which 0.15 mL of analytes were added to 0.5 mL of a sample of lubricating eye drops. Spike-and-recovery and linearity-of-dilution experiments are important methods for validating and assessing the accuracy of ICP OES. Spike and recovery are used to determine whether analyte detection is affected by differences in the standard curve diluent and organic sample matrix.

4.5. Calibration Curves

Elementary standard stock solutions of 100 mg/L of Al, Co, Ca, Cr, Cu, Fe, K, Mg, Mn, Na, Ni, P, Se, and Zn (Specsol, São Paulo, Brazil) were utilized. In addition, calibration curves for all analytes were obtained using eleven different concentrations in the range from 0.001 ppm to 2 ppm.
The LOD and LOQ followed the analytical standards established by the IUPAC [45]. For each element detected, the LOD, LOQ, and R2 values were determined.

4.6. Determination of the Maximum Daily Dose of Lubricating Eye Drops

For the maximum daily dose (mL/day) of lubricating eye drop instilled in the eyes of an adult person, it was considered that it could be obtained through multiplying the volume of 2 drops (equivalent to 1 drop in each eye) by the maximum frequency of daily instillation (5 times a day) (See Table 5).

4.7. Comparative Study

Upon instillation, part of the eye drops on the ocular surface is absorbed by the cornea, conjunctiva, and sclera, and most of it goes to the systemic circulation via the mucous membrane and rhinopharynx. That is, only a small percentage of the applied dose is released into the intraocular tissues, while about 50–100% of the administered dose may reach the systemic circulation via the conjunctiva and nasolacrimal duct [98,99,100].
Parenterally administered drugs also enter the systemic circulation directly and are not subjected to the first-pass effect or the gastrointestinal environment. Therefore, concentrations of metals in the eye drops, as well as the daily application, can be compared with the values of concentrations established by the Brazilian Pharmacopoeia (BP) and ICH Q3D guideline (R2) for the parenteral route.

4.7.1. Permissible Concentrations of Elemental Impurities in Lubricating Eye Drops

Since the elemental impurities (EIs) absorbed through the ophthalmic route are not considered in the pharmacopoeias and in the ICH Q3D (R2) guideline [40,41,43], in our study, the concentrations of heavy metals and metalloids quantified in the samples of lubricating eye drops were compared with the values of allowed concentrations of EIs in parenteral drugs (µg/g) established by the BP and ICH Q3D guideline (R2).

4.7.2. PDE to Elemental Impurities in Ophthalmic Medications

The PDE in micrograms per day (µg/day) give the maximum permitted quantity of each element that may be, in a medication, administered through the oral, inhaled, and parenteral routes [40,41,43]. However, there are no reference values for comparing the PDE in the ophthalmic pathway.
Therefore, in our study, the daily exposure to elemental impurities (EIs) through the instillation of lubricating eye drops in the eyes (µg/day) was compared to the PDEs for parenteral drugs defined in the ICH Q3D guideline (R2). In addition, the ICH Q3D (R2) guideline allows for the use of parenterally administered PDE values without modification [40,41,42].

4.8. Statistical Analysis

One-way analysis of variance (ANOVA) and Tukey’s post hoc multiple comparison test were used to test for significant differences in concentrations between eye drop brands and significant differences in values of daily instillation of lubricating eye drops with and without preservatives. All statistical analyzes were performed using the GraphPad Prism 9.0 statistical package (San Diego, CA, United States). The significance level was set at p < 0.05.

5. Conclusions

In the present study, samples of 19 brands of Brazilian eye drops used to treat dry eye with and without preservatives showed different concentration values of As, Ba, Cd, Co, Cu, Mn, Mo, Ni, Se, V, Zn, Al, Cr, Fe, Mn, and Pb.
There are no concentration limit values for Al, Se, Fe, Mg, Ba, Co, and Se in drugs established by the BP and Mn defined by the ICH Q3D guideline (R2) for the parenteral route. However, the contents of As in the eye drops with preservatives (four different samples) and one eye drop without preservatives, as well as Cd in three eye drops with preservatives are all above the concentration value defined in the BP for parenteral use. One eye drop with preservatives had a Co concentration value greater than that established by the ICH Q3D guideline (R2) parenterally. On the other hand, the concentrations of Ba, Cr, Cu, Mn, Mo, and Ni in the eye drops were below the values established by the BP or set by the ICH Q3D (R2) parenteral guideline.
The concentrations of As, Ba, Co, Cd, Cr, Cu, Mo, Ni, V, Pb, and Se in some samples of eye drops was below the limits of the PDE set by the ICH Q3D guideline (R2).
In several countries, regulatory agencies must carry out the inspection of eye drops used in the treatment of ophthalmic diseases. Therefore, complementary studies are needed to investigate the possible risks of toxicity due to the daily instillation of lubricating eye drops containing heavy metals and metalloids.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules28186508/s1, Table S1: Statistical test used in view of the results of Table 2 on the concentration of quantified metal(loid)s in lubricating eye drops; Table S2: Statistical test used in view of the results of Table 3 on values of the daily instillation of lubricating eye drops.

Author Contributions

Conceptualization, M.d.O., I.V.S., M.P.H. and L.F.T.M.; methodology, E.S.d.P.M., C.M.L.C., A.F. and M.d.O.; software, M.d.O.; validation, E.S.d.P.M. and M.d.O.; formal analysis, M.d.O., E.S.d.P.M. and V.A.d.N.; investigation, M.d.O., V.T.B. and T.C.d.S.; resources, E.S.d.P.M., V.A.d.N. and M.d.O.; data curation, M.d.O.; writing—original draft preparation, M.d.O.; writing—review and editing, V.A.d.N. and M.d.O.; visualization, M.d.O. and V.A.d.N.; project administration, V.A.d.N.; supervision, V.A.d.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research was financially supported by the Brazilian Research Council (CNPq) (CNPq: Process No 310621/2020-8) and the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior-Brasil (CAPES)-Finance Code 001.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data used to support the findings of this study are available from the corresponding author upon request.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Not applicable.

References

  1. WHO. World Report on Vision; World Health Organization: Geneva, Switzerland, 2019; ISBN 978-92-4-151657-0. [Google Scholar]
  2. Tchegnon, E.; Liao, C.-P.; Ghotbi, E.; Shipman, T.; Wang, Y.; McKay, R.M.; Le, L.Q. Epithelial Stem Cell Homeostasis in Meibomian Gland Development, Dysfunction, and Dry Eye Disease. JCI Insight 2021, 6, e151078. [Google Scholar] [CrossRef]
  3. Golden, M.I.; Meyer, J.J.; Patel, B.C. Dry Eye Syndrome; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  4. Şimşek, C.; Doğru, M.; Kojima, T.; Tsubota, K. Current Management and Treatment of Dry Eye Disease. Turk. J. Ophthalmol. 2018, 48, 309–313. [Google Scholar] [CrossRef] [PubMed]
  5. Kannan, R.; Das, S.; Shetty, R.; Zhou, L.; Ghosh, A.; Deshpande, V. Tear Proteomics in Dry Eye Disease. Indian J. Ophthalmol. 2023, 71, 1203–1214. [Google Scholar] [CrossRef]
  6. Wolffsohn, J.S.; Arita, R.; Chalmers, R.; Djalilian, A.; Dogru, M.; Dumbleton, K.; Gupta, P.K.; Karpecki, P.; Lazreg, S.; Pult, H.; et al. TFOS DEWS II Diagnostic Methodology Report. Ocul. Surf. 2017, 15, 539–574. [Google Scholar] [CrossRef] [PubMed]
  7. Messmer, E.M. The Pathophysiology, Diagnosis, and Treatment of Dry Eye Disease. Dtsch. Arztebl. Int. 2015, 112, 71–82. [Google Scholar] [CrossRef]
  8. Fonseca, E.C.; Arruda, G.V.; Rocha, E.M. Dry eye: Etiopathogenesis and treatment. Arq. Bras. Oftalmol. 2010, 73, 197–203. [Google Scholar] [CrossRef] [PubMed]
  9. Jones, L.; Downie, L.E.; Korb, D.; Benitez-del-Castillo, J.M.; Dana, R.; Deng, S.X.; Dong, P.N.; Geerling, G.; Hida, R.Y.; Liu, Y.; et al. TFOS DEWS II Management and Therapy Report. Ocul. Surf. 2017, 15, 575–628. [Google Scholar] [CrossRef] [PubMed]
  10. Gomes, J.A.P.; Azar, D.T.; Baudouin, C.; Efron, N.; Hirayama, M.; Horwath-Winter, J.; Kim, T.; Mehta, J.S.; Messmer, E.M.; Pepose, J.S.; et al. TFOS DEWS II Iatrogenic Report. Ocul. Surf. 2017, 15, 511–538. [Google Scholar] [CrossRef]
  11. Goldstein, M.H.; Silva, F.Q.; Blender, N.; Tran, T.; Vantipalli, S. Ocular Benzalkonium Chloride Exposure: Problems and Solutions. Eye 2022, 36, 361–368. [Google Scholar] [CrossRef] [PubMed]
  12. Baudouin, C.; Labbé, A.; Liang, H.; Pauly, A.; Brignole-Baudouin, F. Preservatives in Eyedrops: The Good, the Bad and the Ugly. Prog. Retin. Eye Res. 2010, 29, 312–334. [Google Scholar] [CrossRef]
  13. Lee, S.H.; Kang, E.M.; Kim, G.A.; Kwak, S.W.; Kim, J.M.; Bae, H.W.; Seong, G.J.; Kim, C.Y. Three Toxic Heavy Metals in Open-Angle Glaucoma with Low-Teen and High-Teen Intraocular Pressure: A Cross-Sectional Study from South Korea. PLoS ONE 2016, 11, e0164983. [Google Scholar] [CrossRef]
  14. Bocca, B.; Forte, G.; Pisano, A.; Farace, C.; Giancipoli, E.; Pinna, A.; Dore, S.; Madeddu, R. A Pilot Study to Evaluate the Levels of Aqueous Humor Trace Elements in Open-Angle Glaucoma. J. Trace Elem. Med. Biol. 2020, 61, 126560. [Google Scholar] [CrossRef]
  15. Park, S.J.; Lee, J.H.; Woo, S.J.; Kang, S.W.; Park, K.H. Five Heavy Metallic Elements and Age-Related Macular Degeneration: Korean National Health and Nutrition Examination Survey, 2008–2011. Ophthalmology 2015, 122, 129–137. [Google Scholar] [CrossRef]
  16. Jünemann, A.G.M.; Stopa, P.; Michalke, B.; Chaudhri, A.; Reulbach, U.; Huchzermeyer, C.; Schlötzer-Schrehardt, U.; Kruse, F.E.; Zrenner, E.; Rejdak, R. Levels of Aqueous Humor Trace Elements in Patients with Non-Exsudative Age-Related Macular Degeneration: A Case-Control Study. PLoS ONE 2013, 8, e56734. [Google Scholar] [CrossRef]
  17. Aberami, S.; Nikhalashree, S.; Bharathselvi, M.; Biswas, J.; Sulochana, K.N.; Coral, K. Elemental Concentrations in Choroid-RPE and Retina of Human Eyes with Age-Related Macular Degeneration. Exp. Eye Res. 2019, 186, 107718. [Google Scholar] [CrossRef]
  18. Dolar-Szczasny, J.; Święch, A.; Flieger, J.; Tatarczak-Michalewska, M.; Niedzielski, P.; Proch, J.; Majerek, D.; Kawka, J.; Mackiewicz, J. Levels of Trace Elements in the Aqueous Humor of Cataract Patients Measured by the Inductively Coupled Plasma Optical Emission Spectrometry. Molecules 2019, 24, 4127. [Google Scholar] [CrossRef] [PubMed]
  19. Garner, B.; Roberg, K.; Qian, M.; Eaton, J.W.; Truscott, R.J.W. Distribution of Ferritin and Redox-Active Transition Metals in Normal and Cataractous Human Lenses. Exp. Eye Res. 2000, 71, 599–607. [Google Scholar] [CrossRef] [PubMed]
  20. Liou, Y.; Chen, Y.; Chen, W.; Li, K.; Chou, T.; Huang, Y.; Wang, C.; Lai, C. Associations between Biomarkers of Metal Exposure and Dry Eye Metrics in Shipyard Welders: A Cross-Sectional Study. Int. J. Environ. Res. Public Health 2022, 19, 2264. [Google Scholar] [CrossRef]
  21. Jung, S.J.; Lee, S.H. Association between Three Heavy Metals and Dry Eye Disease in Korean Adults: Results of the Korean National Health and Nutrition Examination Survey. Korean J. Ophthalmol. 2019, 33, 26–35. [Google Scholar] [CrossRef]
  22. Chung, S.; Myong, J. Are Higher Blood Mercury Levels Associated with Dry Eye Symptoms in Adult Koreans? A Population-Based Cross-Sectional Study. BMJ Open 2016, 6, e010985. [Google Scholar] [CrossRef]
  23. Choi, M.R.; Ju, M.J.; Kim, D.H.; Choi, Y. Comparison of Environmental Phthalates and Heavy Metals Exposures by Dry Eye Disease Status. In Proceedings of the Korean Society of Environmental Health Conference Materials, Seoul, Republic of Korea, 13 November 2020; Volume 23, p. 142. [Google Scholar]
  24. Chen, Y.; Chen, Y.; Lai, C. Clinical Association between Trace Elements of Tear and Dry Eye Metrics. Sci. Rep. 2022, 12, 18052. [Google Scholar] [CrossRef] [PubMed]
  25. Micun, Z.; Falkowska, M.; Młynarczyk, M.; Kochanowicz, J.; Socha, K.; Konopińska, J. Levels of Trace Elements in the Lens, Aqueous Humour, and Plasma of Cataractous Patients—A Narrative Review. Int. J. Environ. Res. Public Health 2022, 19, 10376. [Google Scholar] [CrossRef] [PubMed]
  26. Cekic, O. Effect of Cigarette Smoking on Copper, Lead, and Cadmium Accumulation in Human Lens. Br. J. Ophthalmol. 1998, 82, 186–188. [Google Scholar] [CrossRef]
  27. Ekinci, M.; Ceylan, E.; Çağatay, H.H.; Keleş, S.; Altınkaynak, H.; Kartal, B.; Koban, Y.; Hüseyinoğlu, N. Occupational Exposure to Lead Decreases Macular, Choroidal, and Retinal Nerve Fiber Layer Thickness in Industrial Battery Workers. Curr. Eye Res. 2014, 39, 853–858. [Google Scholar] [CrossRef]
  28. Sharma, A.; Jain, S. Elemental Impurities in Drug Product. Int. J. Pharm. Sci. Res. 2017, 2, 31–38. [Google Scholar]
  29. Raghuram, P.; Raghuram, P.; Raju, I.V.S.; Raju, I.V.S.; Sriramulu, J. Heavy Metals Testing in Active Pharmaceutical Ingredients: An Alternate Approach. Die Pharm.-Int. J. Pharm. Sci. 2010, 65, 15–18. [Google Scholar] [CrossRef]
  30. Balaram, V. Recent Advances in the Determination of Elemental Impurities in Pharmaceuticals–Status, Challenges and Moving Frontiers. TrAC Trends Anal. Chem. 2016, 80, 83–95. [Google Scholar] [CrossRef]
  31. BRASIL. Brazilian Pharmacopoeia, 6th ed.; National Health Surveillance Agency: Brasília, Brazil, 2019; Volume 1. [Google Scholar]
  32. Kameyama, Y.; Matsuhama, M.; Mizumaru, C.; Saito, R.; Ando, T.; Miyazaki, S. Comparative Study of Pharmacopoeias in Japan, Europe, and the United States: Toward the Further Convergence of International Pharmacopoeial Standards. Chem. Pharm. Bull. 2019, 67, 1301–1313. [Google Scholar] [CrossRef]
  33. WHO. Index of World Pharmacopoeias and Pharmacopoeial Authorities; World Health Organization: Geneva, Switzerland, 2022. [Google Scholar]
  34. Brazil, N.H.S.A. Implementation of the Addendum to the ICH Good Clinical Practice E6 Guide 2016. August 2017. Available online: http://antigo.anvisa.gov.br/documents/33836/2492430/INFORME+-+Adendo+ICH+E6/f59e4e6d-677b-495c-9b08-1108a637b483?version=1.1 (accessed on 28 July 2023).
  35. EMA ICH Q3D Elemental Impurities. Available online: https://www.ema.europa.eu/en/ich-q3d-elemental-impurities (accessed on 4 November 2022).
  36. CANADA, H. Q3D Notice: Health Canada Recommendations for Implementation of the ICH Harmonised Guideline for Elemental Impurities (Q3D) for New and Marketed Products. Available online: https://www.canada.ca/en/health-canada/services/drugs-health-products/drug-products/applications-submissions/guidance-documents/international-conference-harmonisation/quality/notice-health-canada-recommendations-implementation-harmonised-guideline-elemental-impurities-new-marketed-products.html (accessed on 18 May 2023).
  37. Gupta, V.K.; Ali, I.; Khurana, U.; Dhagarra, N. TLC Separation of Transition Metal Ions and Their Quantitative Estimation by Atomic Absorption Spectroscopy. J. Liq. Chromatogr. 1995, 18, 1671–1681. [Google Scholar] [CrossRef]
  38. Ali, I.; Aboul-Enein, H.Y. Speciation of Metal Ions by Capillary Electrophoresis. Crit. Rev. Anal. Chem. 2002, 32, 337–350. [Google Scholar] [CrossRef]
  39. Ali, I.; Aboul-Enein, H.Y. Determination of Metal Ions in Water, Soil, and Sediment by Capillary Electrophoresis. Anal. Lett. 2002, 35, 2053–2076. [Google Scholar] [CrossRef]
  40. ICH Guideline for Elemental Impurities Q3D (R2); The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human: Geneva, Switzerland, 2022.
  41. Silva, R.F.T. Range and Output Simulation for Elemental Impurities in Drug Products. Master Thesis, University of Lisbon, Lisbon, Portugal, 2018. [Google Scholar]
  42. Sanderson, J.; Whitecotton, L. Analysis of Artificial Tear Eye Drops for Elemental Impurities. Using an ICP-MS and USP <232>/<233> and ICH Q3D(R2)/Q2(R1) Protocols. Agilent Technologies, Inc. 2022. Available online: https://www.agilent.com/cs/library/applications/application-elemental-impurity-sated-7900-icp-ms-5994-1561en-agilent.pdf (accessed on 28 July 2023).
  43. USP <232>-Elemental Impurities-Limits. In First Supplement to USP 40–NF 35; The United States Pharmacopeial Convention: Rockville, MD, USA, 2017.
  44. Geronimo, A.C.R.; Melo, E.S.P.; Silva, K.R.N.; Pereira, H.S.; Nascimento, V.A.; Machate, D.J.; do Nascimento, V.A. Human Health Risk Assessment of Heavy Metals and Metalloids in Herbal Medicines Used to Treat Anxiety: Monitoring of Safety. Front. Pharmacol. 2021, 12, 772928. [Google Scholar] [CrossRef]
  45. Long, G.L.; Winefordner, J.D. Limit of Detection. A Closer Look at the IUPAC Definition. Available online: https://pubs.acs.org/doi/pdf/10.1021/ac00258a001 (accessed on 7 November 2022).
  46. Becaria, A.; Campbell, A.; Bondy, S. Aluminum as a Toxicant. Toxicol. Ind. Health 2002, 18, 309–320. [Google Scholar] [CrossRef] [PubMed]
  47. Lu, Z.-Y.; Gong, H.; Amemiya, T. Aluminum Chloride Induces Retinal Changes in the Rat. Toxicol. Sci. 2002, 66, 253–260. [Google Scholar] [CrossRef] [PubMed]
  48. Jomova, K.; Jenisova, Z.; Feszterova, M.; Baros, S.; Liska, J.; Hudecova, D.; Rhodes, C.J.; Valko, M. Arsenic: Toxicity, Oxidative Stress and Human Disease. J. Appl. Toxicol. 2011, 31, 95–107. [Google Scholar] [CrossRef] [PubMed]
  49. Das, D.; Bindhani, B.; Mukherjee, B.; Saha, H.; Biswas, P.; Dutta, K.; Prasad, P.; Sinha, D.; Ray, M.R. Chronic Low-Level Arsenic Exposure Reduces Lung Function in Male Population without Skin Lesions. Int. J. Public Health 2014, 59, 655–663. [Google Scholar] [CrossRef] [PubMed]
  50. Barra, C.M.; Santelli, R.E.; Abrão, J.J.; de la Guardia, M. Especiação de Arsênio-uma revisão. Quím. Nova 2000, 23, 58–70. [Google Scholar] [CrossRef]
  51. Freund, P.; Al-Shafai, L.; Mankovskii, G.; Howarth, D.; Margolin, E. Clinicopathological Correlates: Chronic Arsenic Toxicity Causing Bilateral Symmetric Progressive Optic Neuropathy. J. Neuroophthalmol. 2020, 40, 423–427. [Google Scholar] [CrossRef]
  52. Oskarsson, A. Chapter 4—Barium. In Handbook on the Toxicology of Metals, 5th ed.; Nordberg, G.F., Costa, M., Eds.; Academic Press: Cambridge, MA, USA, 2022; pp. 91–100. ISBN 978-0-12-822946-0. [Google Scholar]
  53. Savazzi, E.A. Determination of the Presence of Barium, Lead and Chromium in Groundwater Samples Collected from the Bauru Aquifer. Master’s Thesis, University of Sao Paulo, Sao Paulo, Brazil, 2009. [Google Scholar]
  54. Young, J.A. Barium Nitrate. J. Chem. Educ. 2006, 83, 34. [Google Scholar] [CrossRef]
  55. Matsushita, K.; Kawashima, R.; Hashida, N.; Hamano, Y.; Harada, K.; Higashisaka, K.; Baba, K.; Sato, S.; Huang, W.; Matsumoto, H.; et al. Barium-Induced Toxic Anterior Segment Syndrome. Eur. J. Ophthalmol. 2021, 33, NP31–NP35. [Google Scholar] [CrossRef]
  56. WHO. International Programme on Chemical Safety. In Barium: Health and Safety Guide; Health and safety guide; World Health Organization: Geneva, Switzerland, 1991; ISBN 978-92-4-151046-2. [Google Scholar]
  57. Volpe, M.G.; Nazzaro, M.; Coppola, R.; Rapuano, F.; Aquino, R.P. Determination and Assessments of Selected Heavy Metals in Eye Shadow Cosmetics from China, Italy, and USA. Microchem. J. 2012, 101, 65–69. [Google Scholar] [CrossRef]
  58. The International Agency for Research on Cancer. Chlorinated Drinking-Water, Chlorination by-Products, Some Other Halogenated Compounds, Cobalt and Cobalt Compounds. In Proceedings of the Chlorinated Drinking-Water, Chlorination By-Products, Some Other Halogenated Compounds, Cobalt and Cobalt Compounds; International Agency for Research on Cancer: Lyon, France, 1991.
  59. Garcia, M.D.; Hur, M.; Chen, J.J.; Bhatti, M.T. Cobalt Toxic Optic Neuropathy and Retinopathy: Case Report and Review of the Literature. Am. J. Ophthalmol. Case Rep. 2020, 17, 100606. [Google Scholar] [CrossRef]
  60. Vennam, S.; Georgoulas, S.; Khawaja, A.; Chua, S.; Strouthidis, N.G.; Foster, P.J. Heavy Metal Toxicity and the Aetiology of Glaucoma. Eye 2020, 34, 129–137. [Google Scholar] [CrossRef] [PubMed]
  61. Ng, S.K.; Ebneter, A.; Gilhotra, J.S. Hip-Implant Related Chorio-Retinal Cobalt Toxicity. Indian J. Ophthalmol. 2013, 61, 35–37. [Google Scholar] [CrossRef] [PubMed]
  62. Lim, C.A.; Khan, J.; Chelva, E.; Khan, R.; Unsworth-Smith, T. The Effect of Cobalt on the Human Eye. Doc. Ophthalmol. 2015, 130, 43–48. [Google Scholar] [CrossRef]
  63. Lozi, A.A. Comparative Toxicity of Heavy Metals, Arsenic, Cadmium, Lead, Chromium and Nickel, on Reproductive Parameters of Adult Male Mice after Acute Exposure. Master Thesis, Federal University of Viçosa, Viçosa, Brazil, 2019. [Google Scholar]
  64. Silva, M.F.d.J. Assessment of health risks resulting from mixtures of metals potentially present in medicines. In Integrated Master in Pharmaceutical Sciences; Higher Institute of Health Sciences Egas Moniz: Caparica, Portugal, 2017. [Google Scholar]
  65. Kaparwan, A. Hexavalent Chromium Induced Toxicity in Nature and Living Beings. Int. Educ. Sci. Res. J. 2023, 9, 3–9. [Google Scholar]
  66. Von Burg, R.; Liu, D. Chromium and Hexavalent Chromium. J. Appl. Toxicol. 1993, 13, 225–230. [Google Scholar] [CrossRef]
  67. Waggoner, D.J.; Bartnikas, T.B.; Gitlin, J.D. The Role of Copper in Neurodegenerative Disease. Neurobiol. Dis. 1999, 6, 221–230. [Google Scholar] [CrossRef]
  68. Haddad, N.; Alasadi, S.; Haddad, H. Contamination of Heavy Metals (Lead, Zinc, Magnesium and Manganese) Concentrations in Human Eyes. Am. J. Anal. Chem. 2012, 337065, 491–494. [Google Scholar] [CrossRef]
  69. McGahan, M.C.; Bito, L.Z.; Myers, B.M. The Pathophysiology of the Ocular Microenvironment. II. Copper-Induced Ocular Inflammation and Hypotony. Exp. Eye Res. 1986, 42, 595–605. [Google Scholar] [CrossRef] [PubMed]
  70. Ayenimo, J.G.; Yusuf, A.M.; Adekunle, A.S.; Makinde, O.W. Heavy Metal Exposure from Personal Care Products. Bull. Environ Contam. Toxicol. 2010, 84, 8–14. [Google Scholar] [CrossRef]
  71. Benarroch, E.E. Brain Iron Homeostasis and Neurodegenerative Disease. Neurology 2009, 72, 1436–1440. [Google Scholar] [CrossRef] [PubMed]
  72. Ugarte, M.; Osborne, N.N.; Brown, L.A.; Bishop, P.N. Iron, Zinc, and Copper in Retinal Physiology and Disease. Surv. Ophthalmol. 2013, 58, 585–609. [Google Scholar] [CrossRef]
  73. Bhasin, G.; Kauser, H.; Athar, M. Iron Augments Stage-I and Stage-II Tumor Promotion in Murine Skin. Cancer Lett. 2002, 183, 113–122. [Google Scholar] [CrossRef]
  74. Wong, R.W.; Richa, D.C.; Hahn, P.; Green, W.R.; Dunaief, J.L. Iron Toxicity as a Potential Factor in AMD. Retina 2007, 27, 997–1003. [Google Scholar] [CrossRef]
  75. Ajib, F.A.; Childress, J.M. Magnesium Toxicity; StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
  76. Lei, M.; Zhu, Y.; Liang, S. Toxic effects of manganese ion eyedrops to ocular anterior segment. Health Environ. Res. Online (HERO) 2016, 34, 510–515. [Google Scholar]
  77. Khosla, P.K.; Karki, D.B.; Gahlot, D.K. Retinotoxic Effect of Zinc, Manganese and Molybdenum on Rabbit Retina (an Experimental Study). Indian J. Ophthalmol. 1983, 31, 545–547. [Google Scholar]
  78. Ceylan, O.; Can Demirdöğen, B.; Mumcuoglu, T.; Aykut, O. Evaluation of Essential and Toxic Trace Elements in Pseudoexfoliation Syndrome and Pseudoexfoliation Glaucoma. Biol. Trace Elem. Res. 2013, 153, 28–34. [Google Scholar] [CrossRef]
  79. Ott, H.C.; Prior, C.; Herold, M.; Riha, M.; Laufer, G.; Ott, G. Respiratory Symptoms and Bronchoalveolar Lavage Abnormalities in Molybdenum Exposed Workers. Wien. Klin. Wochenschr. 2004, 116, 25–30. [Google Scholar]
  80. Jonmaire, P. Molybdenum. In Hamilton & Hardy’s Industrial Toxicology; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2015; pp. 167–172. ISBN 978-1-118-83401-5. [Google Scholar]
  81. Mani, P.; Vijayavathi, B.S. Nickel as an Essential Element and a Toxicant. Int. J. Environ. Sci. 2012, 1, 285–288. [Google Scholar]
  82. Nagarjuna, A.; Mohan, D. Biochemical and Histopathological Changes Induced by Nickel in the Striped Mullet, Mugil Cephalus (Linnaeus 1758). Bull. Environ. Contam. Toxicol. 2017, 98, 33–40. [Google Scholar] [CrossRef]
  83. Nelke, K.; Mulak, M.; Łuczak, K.; Pawlak, W.; Nienartowicz, J.; Szumny, D.; Morawska-Kochman, M.; Gerber, H. Occurrence and Exposure to Lead and Cadmium and Their Environmental Influence on Eye Sight. Pol. J. Environ. Stud. 2015, 24, 1491–1496. [Google Scholar] [CrossRef]
  84. Martin, S.; Griswold, W. Human Health Effects of Heavy Metals; Center for Hazardous Substance Research: Manhattan, KS, USA, 2009. [Google Scholar]
  85. ATSDR. Toxicological Profile for Selenium; Agency for Toxic Substances and Disease Registry, U.S.: Atlanta, GA, USA, 2003. [Google Scholar]
  86. Bruhn, R.L.; Stamer, W.D.; Herrygers, L.A.; Levine, J.M.; Noecker, R.J. Relationship between Glaucoma and Selenium Levels in Plasma and Aqueous Humour. Br. J. Ophthalmol. 2009, 93, 1155–1158. [Google Scholar] [CrossRef]
  87. Karaküçük, S.; Mirza, G.E.; Ekinciler, Ö.F.; Saraymen, R.; Karaküçük, İ.; Üstdal, M. Selenium Concentrations in Serum, Lens and Aqueous Humour of Patients with Senile Cataract. Acta Ophthalmol. Scand. 1995, 73, 329–332. [Google Scholar] [CrossRef]
  88. EMEA. Guideline on the Specification Limits for Residues of Metal Catalysts or Metal Reagents; European Medicines Agency: London, UK, 2008. [Google Scholar]
  89. Fallahi, P.; Foddis, R.; Elia, G.; Ragusa, F.; Patrizio, A.; Guglielmi, G.; Frenzilli, G.; Benvenga, S.; Cristaudo, A.; Antonelli, A.; et al. Induction of Th1 Chemokine Secretion in Dermal Fibroblasts by Vanadium Pentoxide. Mol. Med. Rep. 2018, 17, 6914–6918. [Google Scholar] [CrossRef]
  90. Barceloux, D.G.; Barceloux, D. Vanadium. J. Toxicol. Clin. Toxicol. 1999, 37, 265–278. [Google Scholar] [CrossRef]
  91. Cervantes-Yépez, S.; López-Zepeda, L.S.; Fortoul, T.I. Vanadium Inhalation Induces Retinal Müller Glial Cell (MGC) Alterations in a Murine Model. Cutan. Ocul. Toxicol. 2018, 37, 200–206. [Google Scholar] [CrossRef]
  92. Rojas-Lemus, M.; López-Valdez, N.; Bizarro-Nevares, P.; González-Villalva, A.; Ustarroz-Cano, M.; Zepeda-Rodríguez, A.; Pasos-Nájera, F.; García-Peláez, I.; Rivera-Fernández, N.; Fortoul, T.I. Toxic Effects of Inhaled Vanadium Attached to Particulate Matter: A Literature Review. Int. J. Environ. Res. Public Health 2021, 18, 8457. [Google Scholar] [CrossRef]
  93. Vishwanathan, R.; Chung, M.; Johnson, E.J. A Systematic Review on Zinc for the Prevention and Treatment of Age-Related Macular Degeneration. Investig. Ophthalmol. Vis. Sci. 2013, 54, 3985–3998. [Google Scholar] [CrossRef]
  94. Nriagu, J. Zinc Toxicity in Humans; Elsevier B. V.: Amsterdam, The Netherlands, 2007. [Google Scholar]
  95. Amartey, E.O.; Asumadu-Sakyi, A.B.; Adjei, C.A.; Quashie, F.K.; Duodu, G.O.; Bentil, N.O. Determination of Heavy Metals Concentration in Hair Pomades on the Ghanaian Market Using Atomic Absorption Spectrometry Technique. Br. J. Pharmacol. Toxicol. 2011, 2, 192–198. [Google Scholar]
  96. Ezeabara, C.; Ogochukwu, O.; Emeka, A.; Okeke, C.; Mbaekwe, E. Heavy Metal Contamination of Herbal Drugs: Implication for Human Health-A Review. Int. J. Trop. Dis. Health 2014, 4, 1044–1058. [Google Scholar] [CrossRef]
  97. Rai, P.K.; Lee, S.S.; Zhang, M.; Tsang, Y.F.; Kim, K.-H. Heavy Metals in Food Crops: Health Risks, Fate, Mechanisms, and Management. Environ. Int. 2019, 125, 365–385. [Google Scholar] [CrossRef]
  98. Bachu, R.D.; Chowdhury, P.; Al-Saedi, Z.H.F.; Karla, P.K.; Boddu, S.H.S. Ocular Drug Delivery Barriers—Role of Nanocarriers in the Treatment of Anterior Segment Ocular Diseases. Pharmaceutics 2018, 10, 28. [Google Scholar] [CrossRef]
  99. Del Amo, E.M.; Urtti, A. Current and Future Ophthalmic Drug Delivery Systems: A Shift to the Posterior Segment. Drug Discov. Today 2008, 13, 135–143. [Google Scholar] [CrossRef]
  100. Agrahari, V.; Mandal, A.; Agrahari, V.; Trinh, H.M.; Joseph, M.; Ray, A.; Hadji, H.; Mitra, R.; Pal, D.; Mitra, A.K. A Comprehensive Insight on Ocular Pharmacokinetics. Drug Deliv. Transl. Res. 2016, 6, 735–754. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Values of concentrations of metal(loid)s in lubricating eye drops with and without preservatives.
Figure 1. Values of concentrations of metal(loid)s in lubricating eye drops with and without preservatives.
Molecules 28 06508 g001
Figure 2. Values of daily instillation of lubricating eye drops with and without preservatives.
Figure 2. Values of daily instillation of lubricating eye drops with and without preservatives.
Molecules 28 06508 g002
Table 1. Calibration equations (y = ax + b) *, R2, LOD, and LOQ obtained through external calibration.
Table 1. Calibration equations (y = ax + b) *, R2, LOD, and LOQ obtained through external calibration.
ElementCalibration Equations
y = ax + b
LOD (mg/L)LOQ (mg/L)R2
Aly = 5875x − 44.2360.03630.12110.9996
Asy = 449.7x + 1.29770.00360.01210.9996
Bay = 813,519x − 14400.00030.00100.9995
Cdy = 14,392x − 47.2110.00050.00180.9994
Coy = 5674.7x − 0.310.00100.00330.9995
Cry = 17,967x + 30.8120.00110.00370.9994
Cuy = 22,046x + 10.2320.00200.00670.9996
Fey = 10,379x + 18.3520.00650.02170.9994
Mgy = 381,472x + 14390.00170.00570.9994
Mny = 63,173x − 33.5960.00020.00070.9994
Moy = 3475.6x − 5.38570.00060.00190.9993
Niy = 5121.1x − 4.0110.00110.00380.9994
Pby = 995.99x + 9.58880.00500.01680.9993
Sey = 357.79x − 3.05240.00450.01490.9968
Vy = 32,973x − 18.4260.00100.00320.9995
Zny = 10,718x − 37.3130.00160.00520.9989
* y = intensity; a = slope; x = concentration (mg/L); and b = intercept.
Table 2. Concentration of quantified metal(loid)s in lubricating eye drops compared with the permitted concentration of elemental impurities established by the BP [31] and ICH Q3D (R2) guideline for parenteral administration [40].
Table 2. Concentration of quantified metal(loid)s in lubricating eye drops compared with the permitted concentration of elemental impurities established by the BP [31] and ICH Q3D (R2) guideline for parenteral administration [40].
ElementEye Drops 1 * (µg/g)Eye Drops 2 * (µg/g)Eye Drops 3 * (µg/g)Eye Drops 4 * (µg/g)Eye Drops 5 *
(µg/g)
Eye Drops 6 * (µg/g)Eye Drops 7 * (µg/g)BP
(µg/g)
ICH Q3D (R2) (µg/g)
Al0.090 ± 0.020 <LOD<LOD<LOD<LOD<LOD<LODNANA
As0.091 ± 0.006 0.169 ± 0.0090.077 ± 0.0170.088 ± 0.0050.079 ± 0.0070.072 ± 0.0080.108 ± 0.0110.151.5
Ba0.027 ± 0.003 0.024 ± 0.0010.042 ± 0.0010.021 ± 0.0010.021 ± 0.0010.021 ± 0.0010.024 ± 0.002NA70
Cd0.049 ± 0.001 0.051 ± 0.0020.051 ± 0.0020.046 ± 0.0010.047 ± 0.0020.048 ± 0.00050.048 ± 0.00050.050.2
Co0.026 ± 0.004 0.032 ± 0.0020.029 ± 0.0040.014 ± 0.0011.085 ± 0.0190.024 ± 0.0030.025 ± 0.005NA0.5
Cr<LOD<LOD<LOD<LOD<LOD<LOD<LOD2.5110
Cu0.015 ± 0.001 0.012 ± 0.0010.021 ± 0.0020.008 ± 0.0010.004 ± 0.0020.011 ± 0.0140.007 ± 0.0022530
Fe0.167 ± 0.009 <LOD0.170 ± 0.020<LOD<LOD0.022 ± 0.003<LODNANA
Mg0.315 ± 0.008 0.272 ± 0.010.194 ± 0.01<LOD8.224 ± 0.248<LOD5.346 ± 0.162NANA
Mn0.013 ± 0.002 0.012 ± 0.0010.009 ± 0.0010.007 ± 0.0010.006 ± 0.0010.008 ± 0.0010.008 ± 0.00125NA
Mo0.033 ± 0.0020.043 ± 0.0020.032 ± 0.0020.026 ± 0.0010.028 ± 0.0030.029 ± 0.0010.033 ± 0.0022.5150
Ni0.040 ± 0.0030.060 ± 0.0050.030 ± 0.0040.023 ± 0.0020.026 ± 0.0030.028 ± 0.0020.048 ± 0.0022.52
Pb<LOD0.032 ± 0.006<LOD<LOD<LOD<LOD<LOD0.10.5
Se0.245 ± 0.0070.303 ± 0.010.256 ± 0.0130.248 ± 0.0060.245 ± 0.0140.229 ± 0.008 0.261 ± 0.01NA8
V0.014 ± 0.0010.017 ± 0.0050.018 ± 0.0030.014. ± 0.0050.029 ± 0.0030.011 ± 0.004 0.026 ± 0.0072.51
Zn0.246 ± 0.0040.087 ± 0.0030.066 ± 0.0020.052 ± 0.0010.037 ± 0.0020.059 ± 0.0030.065 ± 0.001NANA
ElementEye Drops 8 * (µg/g)Eye Drops 9 * (µg/g)Eye Drops 10 * (µg/g)Eye Drops 11 *
(µg/g)
Eye Drops 12 * (µg/g)Eye Drops 13 * (µg/g)BP
(µg/g)
ICH Q3D (R2) (µg/g)
Al<LOD<LOD<LOD<LOD0.934 ± 0.1091.255 ± 0.034NANA
As0.204 ± 0.0080.195 ± 0.0040.184 ± 0.0040.124 ± 0.0020.147 ± 0.0040.147 ± 0.0080.151.5
Ba0.028 ± 0.0010.031 ± 0.0020.025 ± 0.0010.023 ± 0.0020.025 ± 0.000.023 ± 0.001NA70
Cd0.049 ± 0.0010.051 ± 0.0010.049 ± 0.0010.045 ± 0.0020.049 ± 0.0020.046 ± 0.0020.050.2
Co0.031 ± 0.0020.037 ± 0.0020.029 ± 0.0030.026 ± 0.0070.025 ± 0.0050.019 ± 0.002NA0.5
Cr<LOD0.004 ± 0.001<LOD0.004 ± 0.001<LOD<LOD2.5110
Cu0.015 ± 0.0030.016 ± 0.0020.008 ± 0.0010.003 ± 0.0010.005 ± 0.0020.005 ± 0.0012530
Fe0.013 ± 0.004<LOD<LOD<LOD0.31 ± 0.0220.427 ± 0.026NANA
Mg0.653 ± 0.03624.284 ± 0.7690.489 ± 0.0246.004 ± 0.2245.431 ± 0.1780.258 ± 0.016NANA
Mn0.014 ± 0.0010.014 ± 0.0010.011 ± 0.00030.008 ± 0.0010.01 ± 0.00040.012 ± 0.00125NA
Mo0.043 ± 0.0010.046 ± 0.0010.044 ± 0.0020.035 ± 0.0020.037 ± 0.0010.033 ± 0.0012.5150
Ni0.071 ± 0.0020.065 ± 0.0010.067 ± 0.0030.047 ± 0.0020.056 ± 0.0040.052 ± 0.0042.52
Pb0.019 ± 0.0030.049 ± 0.0110.037 ± 0.0120.009 ± 0.0010.013 ± 0.0060.015 ± 0.00020.10.5
Se0.311 ± 0.0130.358 ± 0.0060.317 ± 0.010.268 ± 0.0140.308 ± 0.0030.299 ± 0.018NA8
V0.018 ± 0.0040.083 ± 0.0050.017 ± 0.0020.022 ± 0.0060.055 ± 0.0040.043 ± 0.0032.51
Zn0.219 ± 0.0010.058 ± 0.0010.186 ± 0.0020.552 ± 0.0080.049 ± 0.0010.200 ± 0.002NANA
ElementEye Drops 14 * (µg/g)Eye Drops 15 ** (µg/g)Eye Drops 16 ** (µg/g)Eye Drops 17 ** (µg/g)Eye Drops 18 ** (µg/g)Eye Drops 19 ** (µg/g)BP
(µg/g)
ICH Q3D (R2) (µg/g)
Al0.073 ± 0.0250.134 ± 0.039<LOD<LOD2.382 ± 0.219<LODNANA
As0.129 ± 0.0010.075 ± 0.0030.178 ± 0.0020.108 ± 0.0090.036 ± 0.0130.122 ± 0.0080.151.5
Ba0.021 ± 0.0010.021 ± 0.0010.039 ± 0.0010.056 ± 0.0020.021 ± 0.0020.03 ± 0.001NA70
Cd0.044 ± 0.0010.049 ± 0.0010.048 ± 0.0020.044 ± 0.0020.047 ± 0.0010.046 ± 0.0010.050.2
Co0.013 ± 0.0010.028 ± 0.0030.021 ± 0.0030.015 ± 0.0040.028 ± 0.0090.031 ± 0.002NA0.5
Cr<LOD<LOD<LOD<LOD<LOD0.020 ± 0.0032.5110
Cu0.002 ± 0.0010.008 ± 0.0030.006 ± 0.0030.023 ± 0.0060.011 ± 0.0090.006 ± 0.0022530
Fe<LOD0.076 ± 0.005<LOD<LOD0.453 ± 0.0320.051 ± 0.011NANA
Mg0.225 ± 0.0090.021 ± 0.007<LOD<LOD0.124 ± 0.014<LODNANA
Mn0.009 ± 0.00030.008 ± 0.00040.007 ± 0.0010.007 ± 0.0010.011 ± 0.0010.008 ± 0.00125NA
Mo0.03 ± 0.0020.027 ± 0.0020.032 ± 0.0020.026 ± 0.0010.025 ± 0.0020.029 ± 0.0012.5150
Ni0.034 ± 0.0010.027 ± 0.0030.043 ± 0.0040.026 ± 0.0040.031 ± 0.0030.03 ± 0.0022.52
Pb<LOD<LOD<LOD<LOD<LOD<LOD0.10.5
Se0.278 ± 0.0110.250 ± 0.0040.365 ± 0.010.277 ± 0.0110.211 ± 0.0090.295 ± 0.001NA8
V0.008 ± 0.0020.014 ± 0.0040.009 ± 0.0040.011 ± 0.0080.026 ± 0.0070.011 ± 0.0032.51
Zn0.034 ± 0.0020.019 ± 0.0020.022 ± 0.0020.024 ± 0.0010.039 ± 0.0020.136 ± 0.003NANA
Notes: data are presented as mean ± standard deviation. * With preservative; ** without preservative; <LOD—analyte concentrations were below detection limits; and NA—not applicable.
Table 3. Values of daily instillation of lubricating eye drops compared with the PDE of ICH Q3D (R2) for parenteral administration [40].
Table 3. Values of daily instillation of lubricating eye drops compared with the PDE of ICH Q3D (R2) for parenteral administration [40].
ElementEye Drops 1 * (µg/day)Eye Drops 2 * (µg/day)Eye Drops 3 * (µg/day)Eye Drops 4 * (µg/day)Eye Drops 5 *
(µg/day)
Eye Drops 6 * (µg/day)PDE of ICH Q3D (R2) for Parenteral Administration (µg/day)
Al0.041<LOD<LOD<LOD<LOD<LODNA
As0.0410.0690.0230.0350.0400.03315
Ba0.012 0.0100.0130.0090.0100.010700
Cd0.0220.0210.0160.0180.0240.0222
Co0.0120.0130.0090.0060.5540.0115
Cr<LOD<LOD<LOD<LOD<LOD<LOD1100
Cu0.0070.0050.0060.0030.0020.005300
Fe0.076<LOD0.052<LOD<LOD0.010NA
Mg0.1430.1110.060<LOD4.194<LODNA
Mn0.0060.0050.0030.0030.0030.003NA
Mo0.0150.0170.0100.0100.0140.0131500
Ni0.0180.0240.0090.0090.0130.01220
Pb<LOD0.013<LOD<LOD<LOD<LOD5
Se0.1110.1240.0780.0990.1250.10480
V0.0070.0070.0050.0060.0150.00510
Zn0.1110.0360.0200.0210.0190.027NA
ElementEye Drops 7 * (µg/day)Eye Drops 8 * (µg/day)Eye Drops 9 * (µg/day)Eye Drops 10 * (µg/day)Eye Drops 11 * (µg/day)Eye Drops 12* (µg/day)PDE of ICH Q3D (R2) for Parenteral
Administration (µg/day)
Al<LOD<LOD<LOD<LOD<LOD0.570NA
As 0.0570.1000.0880.1090.0590.09015
Ba0.0130.0140.0140.0150.0110.015700
Cd0.0250.0240.0230.0290.0210.0302
Co0.013 0.0150.0170.0170.0130.0155
Cr<LOD<LOD0.002<LOD0.002<LOD1100
Cu0.0040.0070.0070.0050.0010.003300
Fe<LOD0.006<LOD<LOD<LOD0.189NA
Mg2.8120.32011.0250.2882.8823.313NA
Mn0.0040.0070.0060.0060.0040.006NA
Mo0.0170.0210.0210.0260.0170.0231500
Ni0.0250.0350.0290.0400.0230.03420
Pb<LOD0.0090.0220.0220.0040.0085
Se0.1370.1520.1630.1870.1280.18880
V0.0140.0090.0380.0100.0100.03410
Zn0.0340.1070.0260.1100.2650.030NA
ElementEye Drops 13 * (µg/day)Eye Drops 14 * (µg/day)Eye Drops 15 ** (µg/day)Eye Drops 16 ** (µg/day)Eye Drops 17 ** (µg/day)Eye Drops 18 ** (µg/day)Eye Drops 19 ** (µg/day)PDE of ICH Q3D (R2) for Parenteral
Administration (µg/day)
Al0.7530.0370.066 <LOD<LOD0.793<LODNA
As0.0880.0660.0370.0360.0220.0120.02415
Ba0.0140.0110.0100.0080.0110.0070.006700
Cd0.0280.022 0.0240.0090.0090.0160.0092
Co0.0110.007 0.0140.0040.0030.0090.0065
Cr<LOD<LOD<LOD<LOD<LOD<LOD0.0041100
Cu0.0030.001 0.0040.0010.0050.0040.001300
Fe0.256<LOD0.037<LOD<LOD0.1510.010NA
Mg0.1550.1150.010<LOD<LOD0.041<LODNA
Mn0.0070.0040.0040.0010.0010.0040.002NA
Mo0.0200.0160.0130.0060.0050.0080.0061500
Ni0.0310.0180.0130.0090.0050.0100.00620
Pb0.009<LOD<LOD<LOD<LOD<LOD<LOD5
Se0.1790.1420.1220.0730.0550.0700.05980
V0.0260.0040.0070.0020.0020.0090.00210
Zn0.1200.018 0.0090.0040.0050.0130.027NA
Notes: data are presented as mean. * With preservative; ** without preservative; <LOD—analyte concentrations were below detection limits; and NA—not applicable.
Table 4. ICP OES instrumental parameters.
Table 4. ICP OES instrumental parameters.
ParameterSetting
RF power1250 W
Sample flow0.45 L/min
Plasma flow rate12 L/min
Integration time5 s
Stabilization time20 s
Nebulization pressure30 psi
Plasma viewAxial
Analytes/λ (nm)Al (309.271); As (189.042); Ba (455.403); Cd (228.802); Co (228.616); Cr (283.563); Cu (324.754); Fe (259.940); Mg (279.553); Mn (257.610); Mo (202.030); Ni (221.647); Pb (220.353); Se (196.090); V (309.311); and Zn (213.856).
Table 5. Lubricating eye drops with (yes) or without (no) preservatives, volume of one drop (mL), and maximum daily frequency of instillations in the eyes and maximum daily dose (mL) (as established in the package inserts).
Table 5. Lubricating eye drops with (yes) or without (no) preservatives, volume of one drop (mL), and maximum daily frequency of instillations in the eyes and maximum daily dose (mL) (as established in the package inserts).
Eye DropsPreservativeVolume: One Drop (mL)Maximum Daily
Frequency
Maximum Daily Dose (Two Eyes) (mL/day)
1Yes0.0455 *0.454
2Yes0.05140.408
3Yes0.05130.306
4Yes0.04050.400
5Yes0.05150.510
6Yes0.0455 *0.454
7Yes0.0535 *0.526
8Yes0.0495 *0.490
9Yes0.0455 *0.454
10Yes0.0595 *0.589
11Yes0.0485 *0.480
12Yes0.0615 *0.610
13Yes0.0605 *0.600
14Yes0.0515 *0.510
15No0.0495 *0.490
16No0.03330.200
17No0.03330.200
18No0.04240.333
19No0.03330.200
* The lubricating eye drop package insert does not contain the maximum daily frequency of instillations in the eyes.
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

Oliveira, M.d.; Melo, E.S.d.P.; Silva, T.C.d.; Cardozo, C.M.L.; Siqueira, I.V.; Hamaji, M.P.; Braga, V.T.; Martin, L.F.T.; Fonseca, A.; Nascimento, V.A.d. Quantification of Metal(loid)s in Lubricating Eye Drops Used in the Treatment of Dry Eye Disease. Molecules 2023, 28, 6508. https://doi.org/10.3390/molecules28186508

AMA Style

Oliveira Md, Melo ESdP, Silva TCd, Cardozo CML, Siqueira IV, Hamaji MP, Braga VT, Martin LFT, Fonseca A, Nascimento VAd. Quantification of Metal(loid)s in Lubricating Eye Drops Used in the Treatment of Dry Eye Disease. Molecules. 2023; 28(18):6508. https://doi.org/10.3390/molecules28186508

Chicago/Turabian Style

Oliveira, Marcelo de, Elaine S. de Pádua Melo, Thaís Carvalho da Silva, Carla Maiara Lopes Cardozo, Igor Valadares Siqueira, Mariana Pereira Hamaji, Vanessa Torres Braga, Luiz Fernando Taranta Martin, Alessandro Fonseca, and Valter Aragão do Nascimento. 2023. "Quantification of Metal(loid)s in Lubricating Eye Drops Used in the Treatment of Dry Eye Disease" Molecules 28, no. 18: 6508. https://doi.org/10.3390/molecules28186508

Article Metrics

Back to TopTop