Next Article in Journal
Characterization and Polydispersity of Volcanic Ash Nanoparticles in Synthetic Lung Fluid
Previous Article in Journal
Air, Dermal, and Urinary Metabolite Levels of Backpack and Tractor Sprayers Using the Herbicide Acetochlor in Thailand
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Determinants of Pesticide Exposure in Occupational Studies: A Meta-Analysis

1
Center for Primary Care and Public Health (Unisanté), Department of Occupational and Environmental Health (DSTE), University of Lausanne, 1066 Epalinges-Lausanne, Switzerland
2
International Labour Organization (ILO), 1211 Geneva, Switzerland
*
Author to whom correspondence should be addressed.
Toxics 2023, 11(7), 623; https://doi.org/10.3390/toxics11070623
Submission received: 13 June 2023 / Revised: 13 July 2023 / Accepted: 15 July 2023 / Published: 18 July 2023
(This article belongs to the Special Issue Human Biomonitoring: Development and Results)

Abstract

:
Few epidemiological studies use exposure determinants specifically tailored to assess pesticide or plant protection product (PPP) exposures when assessing presumed association between occupational exposure and health outcomes among agricultural workers. This lack of exposure specificity could lead to results that fail to detect an association. It could be related to the lack of consensus on exposure assessment methods and the choice of exposure determinants. We conducted a meta-analysis following the PRISMA checklist to identify PPP exposure determinants used in occupational studies and identified exposure determinants that best characterized agricultural exposures to PPPs. Out of 1436 studies identified, 71 were included. The exposure determinants identified were active ingredients, chemical classes, types of PPP, crops, tasks, frequencies, duration, lifetime exposure days, and intensity-weighted exposure days. Only six over 17 associations between exposure determinants and health outcomes were found with moderate quality of evidence. Overall, epidemiological studies had difficulty defining relevant determinants to characterize PPP exposures for agricultural workers. We recommend that a standardized list of determinants for PPP exposures in occupational exposure studies should include information on formulations, intensity, duration, and frequency of PPP exposure. Harmonized data collection on exposure and health outcomes are required as well as standard units for each exposure determinant.

1. Introduction

Occupational risk assessment is an evaluation of the likelihood that an adverse health effect induced by an event, such as Plant Protection Product (PPP) exposure during agricultural tasks, would occur [1]. In these assessments, PPP exposure information is combined with the hazard that derives from the active ingredient. Coformulants in the PPP formulation are not considered. Agricultural workers performing PPP application-related tasks may be exposed to PPPs. PPP exposure depends on the type of activity (e.g., mixing, loading, applying, re-entering, harvesting), application method (e.g., backpack sprayer; broadcast from farm vehicle; air sprayer), use of personal protective equipment (PPE), and other parameters [2,3]. Depending on the route of entry of a given substance, i.e., via skin, ingestion or inhalation [4], its effects may vary. These parameters lead to heterogeneous exposure of workers, which makes risk assessment challenging.
Exposure to PPPs may cause adverse health effects such as neurological diseases, cancer, endocrine disorders, or reproductive disorders [5]. However, there are contrary conclusions regarding a presumed link between PPP exposure and health outcomes in epidemiological studies [6]. We believe that this inconsistency might be due to the lack of exposure assessment as well as disease ascertainment.
Exposure determinants are factors that predict possible exposures to a given substance [7]. Using a set of exposure determinants specifically developed for PPP exposures could refine exposure assessment and consequently, have a better chance of detecting a possible association between exposures and health outcomes in epidemiological studies. The standard method to assess occupational exposure group is the Similar Exposure Group (SEG) approach [8,9]. The SEG approach is widely used in industrial settings to assess possible inhalation exposure. However, it is not often used to characterize exposure for agricultural workers, since skin has always been a major route of exposures among agricultural workers. The validation of SEG is performed using air sampling. However, the main exposure route in agricultural workers is skin exposure (around 90% of exposure) followed by inhalation (around 10% of exposure) [4,10]. Moreover, workers are exposed to different formulations of products (the active ingredient and other ingredients like formulants) [11]. The different formulations can have a different kinetic effect.
Several epidemiological studies evaluating the potential link between exposure to PPPs and health outcomes are ambiguous and the reason might be that exposures to PPPs are rarely characterized [12]. Exposures were often not evaluated on exposure intensity, frequency, and duration. For example, an agricultural worker prepares and manually sprays the product over many days and another worker who does not need to prepare the product and whose sprayer is hooked up to the tractor that sprays for some hours would be classified in the same exposed group. This leads to misclassification and a bias toward the null [13]. Exposure determinants were used in a few epidemiological studies to assess exposures; however, there was no consensus on the choice of exposure determinants nor assessment methods (e.g., self-reported questionnaire, interview).
In this systematic review and meta-analysis, our objectives were to evaluate exposure determinants identified in the occupational epidemiological studies and recommend the most relevant based on their ability to characterize agricultural PPP exposures according to the quality of evidence of the association between exposure determinants and health outcomes. A focus is on the estimate of the association of the exposure determinant with the health outcome.

2. Methods

2.1. Protocol and Registration

This review protocol is available on the international database PROSPERO (Registration number: CRD42022293243). This systematic review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) methodology [14] (see PRISMA checklist in the Supplementary Information).

2.2. Information Sources

We performed a systematic literature search in three databases: Web of Science (maintained by Clarivate analytics), MEDLINE via PubMed, and Scopus, in July 2022. In order to be exhaustive and not exclude changes over time, the search included all articles published from 1st January 1990 to 18th July 2022. Search strings are available in Supplementary Information (Supplementary text S1).

2.3. Eligibility Criteria

The inclusion criteria of studies for the systematic review were (a) the exposed population was agricultural workers; (b) the investigated health outcome was neurological, carcinogenic, mutagenic, reprotoxic (CMR), or endocrine disruption (included thyroid, diabetes); (c) ascribed exposure levels or measured exposures during PPP use; (d) English language; and (e) original research articles. Reviews, meta-analyses, letters to the editor and monographs were excluded. Likewise, studies that did not report health effects or focused on poisoning events (i.e., acute exposure) were excluded. All the papers reporting association using ever/never use categories were excluded.
Title, abstract, and keywords were used for a first-step screening of eligibility using the online tool RAYYAN [15]. After the first screening, articles were read to include only studies using exposure determinants for assessing PPP exposures. Two reviewers (C.O and A.B.) performed the screening and selection of articles independently. At least 10% of the articles were randomly selected and cross-checked. No automated tools were used, except the tool for titles and abstracts screening for keywords and exclusion criteria, which was performed using RAYYAN.

2.4. Data Extraction

Data was extracted from the articles that met the eligibility criteria after screening the abstract and the full text. An Excel file was used to extract relevant data from each publication and compile the following information: Authors’ names, Title, Publication year, Study design (e.g., epidemiologic, case control, cross-sectional, etc.), Health outcome type, Health outcome assessment type (e.g., clinical exams, registers, medical questionnaires, etc.), Study location (country), Crop type, Study period, Study population (n), and Active substances (Table S1, in the supplementary information). Exposure determinants were classified into three categories: sociodemographic data, general farming information, and PPP use practices. The relevance of this classification is discussed in the results’ section. All the results from this systematic review were analyzed for each health outcome independently. Risk estimates related to cancer were combined for all cancer sites in order to increase the statistical power of the results.

2.5. Quality Assessment and Grading

2.5.1. Meta-Analysis

We grouped studies depending on the outcome measured such as cancer, neurotoxicity, or endocrine disruption (diabetes and thyroid disruption). We further divided these four main groups into subgroups based on exposure determinants (e.g., active ingredient, chemical class, etc.). We performed the meta-analysis on the reported ratio (hazard risk (HR), odds ratio (OR), or relative risk (RR)) of the associations between the level in the exposure determinants groups and health outcomes. For example, in one study [16], they reported that the higher users (intensity-weighted lifetime days) of the herbicide 2,4,5-T had an increased risk of renal cell carcinoma (RR = 2.9 (95% CI: 1.65–5.17)) compared to the never users. To facilitate comparison, we converted the reported ratios (HR, OR, or RR) from the included studies into the most commonly reported ratios (HR, OR, or RR) when there were variations. This computation was performed using the formula published in studies [17,18], which takes into account the sample size and the reference rate. We used a restricted maximization likelihood model with the study ID as a random effect. We reported the summary estimates and heterogeneity I2 for the overall meta-analysis and subgroup analyses. In the meta-analysis, a summary estimate was obtained by combining the individual effect estimates (in this review RR or OR) observed in the original included studies that make up the analysis [19]. The I2 statistic quantifies the amount of variation in a meta-analysis that can be attributed to differences between trials rather than random sampling error. Mathematically, I2 is calculated as I2 = τ2/(σ2 + τ2), where τ2 represents between-trial heterogeneity, σ2 represents the shared sampling error across trials, and σ2 + τ2 represent the overall variation in the meta-analysis. Typically, I2 is derived from the formula (Q − df)/Q × 100%, where Q is the statistic for Cochran’s homogeneity test and df is the degree of freedom (equal to the number of trials minus one). Higgins et al. investigated various approaches to determine 95% confidence intervals for the I2 estimate [20]. The threshold to interpret I2 can be misleading due to the several factors that influence the inconsistency. Above 75%, there is considerable heterogeneity [20]. The publication bias was assessed using funnel plots when there were at least 10 studies in a subgroup [20]. Sensitivity analysis was performed by eliminating one study at a time and assessing the effect of this elimination by comparing the summary estimates and the heterogeneity (I2) before and after the elimination of each study [21].

2.5.2. Risk of Bias and Quality Assessment

We used the Joanna Briggs Institute (JBI) checklist for prevalence studies to assess the risk of bias in each article [22,23]. The JBI’s critical appraisal tools help to determine the reliability, relevance, and outcomes of published articles. This checklist provides observations on internal and external validities on the methodological quality of the studies. The overall risk of bias for each study was categorized as either high risk of bias (i.e., more than (and equal to) five criteria out of nine rated “unclear” or “missing”) or low risk of bias (i.e., fewer than five criteria out of nine rated “unclear” or “missing”). The risk of bias was completed for included articles by the lead author (CO), and a second reviewer (AB) independently evaluated 10% of the articles selected randomly for cross-checking.

2.5.3. Grading and Overall Quality of Evidence

We graded the quality of evidence of the associations between exposure determinants and health outcomes following the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach [24]. Following the GRADE, the quality of evidence could be high, moderate, low, or very low. The overall quality of evidence was assessed for each subgroup by evaluating the risk of bias, inconsistency, indirectness, imprecision, and publication bias. Risk of bias being already assessed by JBI, we assessed the remaining criteria. Inconsistency describes how consistent the effects are across included studies. Indirectness compares differences in intervention (exposure in our case), outcome measurements, or population in the studies. For example, there were differences between the population of interest and those who participated in relevant studies. Our systematic review included only farmers who met our eligibility criteria; hence, the evidence regarding farmers was direct by definition. However, in some cases, there could be exceptions and differences between the population of interest and those in the included studies [25]. Imprecision assesses the risk of random errors. We systematically started the assessment by considering the quality of evidence as high and then downgraded it depending on the risk of bias, inconsistency, indirectness, imprecision, and publication bias.

3. Results

3.1. Selected Studies

A total of 1642 records were identified with the literature search in three databases: Web of Science, PubMed, and Scopus (Figure 1). Among them, 206 were duplicates, leaving 1436 articles for the first screening based on titles and abstracts. Following the first screening, 364 articles were excluded because they were not original studies, 43 articles were excluded as they were not published in English and 842 articles were judged to be out of topic. Out of the remaining 187 original articles, 73 were excluded after the full-text screening and 18 were excluded for eligibility (Figure 1). Additionally, 25 studies were excluded from the meta-analysis because they did not report information (i.e., sampling size and population size) to transform the reported ratios into RR to allow comparison among studies (as described in Section 2.5.1). At the end, 71 studies were included in the meta-analysis (Supplementary information, Table S1).
Among the included studies in the systematic review, 37% were published in 2015 or later, 14% between 2010 and 2014, 39% between 2005 and 2009, and 10% between 2000 and 2004. There was no publications before 2000, although studies published after 1990 were included in the systematic review. Over 80% (84.5%) focused on a North American population, followed by a European (14.1%) and an Asian (1.4%) population. The included studies were mainly cohort studies (92%) and some case controls (8%). The majority of the studies (85%) focused on cancer, while endocrine disruption and neurotoxicity health effects made up 8 and 7% of the studies, respectively.
We could not provide a meta-analysis for the two endpoints of mutagenicity and reprotoxicity because the identified studies did not define the outcome in comparable ways. For example, some studies measured reprotoxicity as the number of miscarriages whereas other studies evaluated the sperm quality.
Health outcomes were mostly extracted from registries (82%) or from self-reported questionnaires (11%). Only a few studies integrated a medical doctor in the team to examine participants and diagnose or label their pathologies (3%) or administer psychometric tests to participants (3%). Even fewer studies used biomonitoring (1%).
Among the included studies, 6% were with a low risk of bias, 76% with a moderate risk of bias and 18% with a high risk of bias (Table S2). The low risk of bias articles listed on average several exposure determinants, and the health outcomes were mainly cancer. Most articles with moderate risk of bias did not include a representative sample of the population either because of convenience sampling or an inadequate response rate. Articles with a high risk of bias lacked detailed descriptions of the study subjects, suffered from a lack of information, control groups, or an association measurement.
The Table 1 listed the exposure determinants used to classify participants according to their PPP exposures in the 71 included studies. Twenty-five exposure determinants were identified. Each determinant was defined and assessed in a specific unit of measurement. Reported exposure determinants were very specific in some studies, such as which personal protective equipment (PPE) is used to perform a specific task or more general ones like spraying tasks (Yes or No). Some of the studies combined several exposure determinants to calculate an intensity level (IL) or a cumulative exposure index (CEI) [2]. These measurement tools were implemented initially in the American cohort Agricultural Health Study (AHS) to assess an overall quantitative long-term exposure [2]. The IL and CEI combined different exposure determinants listed in Table 1. A large number of studies considered sociodemographic information such as gender and age as exposure determinants.

3.2. Meta-Analysis Results

The meta-analysis was first performed for each of the four health outcomes (i.e., Cancer: 60 studies; Neurotoxicity: five studies; Endocrine disruptions: thyroid: three studies, diabetes: three studies). Since heterogeneity was high (I2 > 60%) [26], we then divided these disease groups into subgroups by exposure determinants (Table 2). The subgroup analysis provided results with reduced heterogeneity below 30% for the cancer for active ingredient, PPP type, task, frequency, duration, and intensity weighted exposure days as well as for endocrine disruption outcomes and active ingredient and frequency. In some cases, when only two to three studies were included in the subgroup analysis, the heterogeneity equals zero, which means that heterogeneity might not be important [20]. The overall heterogeneity in the subgroup analysis for neurotoxicity was considered moderate (I2 = 43–63%).
The quality of evidence was moderate for the association between two subgroups of exposure determinants with significant results: lifetime exposure days (HR = 0.93, 95%CI = 0.91–0.96, I2 = 40.67), intensity-weighted exposure days (HR = 0.96, 0.95–0.98, I2 = 15.57), and cancer. We found a low quality of evidence for the effect of “task” as an exposure determinant on protecting (HR < 1) from cancer (HR = 0.86, 0.71–0.99) in two studies with heterogeneity I2 = 0. The associations between active ingredient (HR = 1.01, 0.99–1.03), chemical class (HR = 1.05, 0.89–1.2), type of PPP (HR = 1.03, 0.93–1.1), crops (HR = 1.01, 0.98–1.03), frequency (HR = 0.68, 0.08–1.27), and duration (HR = 1.09, 0.99–1.19) with cancer were not significant. We found q low quality of evidence for the type of PPP harmful effect (OR > 1) associated with neurotoxicity (OR = 1.18, 1.11–1.25) in two studies with heterogeneity I2 = 63.44. However, there was no significant association between active ingredient and neurotoxicity (HR = 1.01, 0.98–1.05). There was a low quality of evidence for the association of active ingredient with diabetes (HR = 1.03, 1.01–1.06, I2 = 8.01) in two studies as well as with thyroid disruption (HR = 1.08, 1.05–1.11, I2 = 47.23) in two studies. We found a moderate quality of evidence for the harmful effect of longer duration and diabetes (OR = 1.19, 1.01–1.38) in two studies with heterogeneity I2 = 0. There was no significant association between the frequency and diabetes (HR = 0.94, 0.79–1.08). There was a moderate quality of evidence between harmful effects of intensity-weighted exposure days with thyroid disorder (HR = 1.09, 1.06–1.12) in three studies with heterogeneity I2 = 16.56 (Table 2). Overall, the indirectness had a negative impact and downgraded the quality of evidence for the associations of all subgroups of exposure determinants due to the difference in outcomes assessment, especially for the outcome of neurotoxicity.

3.2.1. Effects of the PPP Exposure Determinants on the Health Outcome Association

Combining several exposure determinants may improve the results of the meta-analysis for the outcome of cancer specifically (Table 3). For example, the heterogeneity decreased for the association between active ingredient and cancer outcome, but the summary estimate was significant when restricting active ingredient with duration (Table 3). This estimate indicated harmful effects of a longer duration of exposure to the active ingredient in developing cancer among exposed farmers.

3.2.2. Publication Bias

The results of publication bias for three groups of determinants (i.e., active ingredient, lifetime exposure days, and intensity-weighted exposure days) associated with cancer are presented in the supplementary material (Supplementary Materials Figures S1–S3). The funnel plots showed an asymmetry in the distribution of studies on both sides of the plots. Consequently, we considered there is evidence for publication bias based on the funnel plots.

4. Discussion

4.1. Main Findings

This systematic review identified 25 exposure determinants (Table 1) in 71 original studies published in the last 30 years. PPP exposure is not comparably assessed in epidemiological studies because they used different exposure determinants. This review highlighted the difficulty in characterizing exposures with specific PPP exposure determinants for agricultural workers. We included only nine exposure determinants in the meta-analysis as these were the only ones evaluated in a minimum of two studies. Two exposure determinants were exposure indices for an intensity level (IL): intensity-weighted exposure days and the cumulative exposure index (CEI) using lifetime exposure days [2]. They combined several exposure determinants. The IL- or CEI-exposure calculation provided a scale to estimate the PPP exposures. The difference between the IL and CEI is that the CEI takes into consideration the duration and frequency of application [27]. As exposure determinants, IL or CEI provided a moderate quality of evidence for their association with cancer and a moderate quality of evidence for their association with thyroid disruption. In our review, 60% of the included studies used a combination of exposure determinants (IL or CEI) to assess the risk of exposure. All these studies used data from the AHS cohort. This approach, using a combination of exposure determinants, could be a tool for epidemiological studies and should be recommended for assessing the overall risk associated with PPP practices. In several other studies, an extensive questionnaire completed by the agricultural workers was used to characterize PPP exposures but not all the data collected were used to assess exposure. Most of the exposure studies reported their results based on a dichotomous category (i.e., exposed and nonexposed). Although, some studies listed exposure determinants in their methodology to assess exposures, they did not necessarily analyze these data [28,29,30]. This emphasized that information requested of participants is not used for assessing PPP exposure even when available.
Out of nine determinants, only lifetime exposure days and intensity-level exposure days had a moderate quality of evidence and a statistically significant association with cancer. This could be explained by the fact that these two determinants combine several exposure determinants (i.e., mixing status, application method, and PPE use). Similarly, the meta-analysis showed that the combination of exposure determinants in the analysis decrease the heterogeneity of the results.

4.2. Results’ Interpretation

Socio-professional data are reported in the selected articles as exposure determinants although they are covariates or adjustment variables. They do not imply any particular effect on the exposure level among agricultural workers [31]. For example, the association between gender and health outcome among agricultural workers has not been evaluated in the literature due to the low number of women applying PPPs. In our meta-analysis, some studies (n = 4) used female gender as one of the exclusion criteria. Likewise, age used as a cumulative value to define exposure duration cannot be considered as a determinant of exposure.
Job title may also induce a bias in exposure determinant when used as a proxy of exposure [28,32]. This exposure misclassification groups people who are assumed to be comparably exposed to the substance of interest based on an individual characteristic, while exposure is an external one [33,34]. Similar conclusions were drawn when job title was used as an exposure assessment method such, as Job Exposure Matrices (JEMs), concerning PPP exposure [35]. Indeed, not all the tasks performed by an operator are considered in the JEMs, possibly leading to an underestimation. For example, few JEMs considered re-entry tasks even though these tasks are suspected of leading to high PPP exposures similar to spraying and mixing tasks [33,36]. However, JEMs are validated tools, more effective for assessing health risks related to occupational exposures than self-reported questionnaires [33] and help to cover a large population at a lower cost [37]. In addition, most of the occupational epidemiological studies evaluated PPP exposures using a self-reported questionnaire [38]. It may be perceived as the easiest and quickest approach to collect information on PPP exposures; however, response and recall bias are known to occur. The main advantage of self-reported questionnaires is that it is a simple way to collect extensive information or data from many participants. Nonetheless, answers may be subjective or invalid depending on the individual’s tendency to respond to questions. This response bias may affect the validity and the reliability of the collected data [39]. Moreover, there is a poor correlation between the estimates from crop-based job-exposure matrices (CEMs) and the self-reported exposure assessment [40]. This can lead to an underestimate of PPP exposure prevalence from self-reported questionnaires. These questionnaires evaluate a qualitative exposure whereas CEMs evaluate semiquantitative or quantitative exposures. In summary, the exposure assessment using proxy or self-reported questionnaire is not accurate and can lead to misclassification.
Biomonitoring, on the contrary, gives quantifiable internal exposure measurements [41]. The use of specific biomarkers and a suitable biological matrix (e.g., hair, urine, blood) are key elements to target exposure [42,43]. Biomonitoring measures the PPP active substance or its metabolites in the body following exposure. Biomonitoring results are the dose absorbed by the workers in a biological matrix independently of the route of exposure, absorption variation, and timing [44,45,46]. Biomonitoring may be a useful tool in epidemiological studies to interpret health-based biological results for short and mid-term effects [47,48]. Biomonitoring can associate the internal dose with exposure determinants. The most relevant exposure determinants can then be included within toxicokinetic models to predict exposures for a larger cohort without collecting samples. Furthermore, biomonitoring results are quantified exposure doses, while questionnaires are exposure estimates prone to response bias. Very few studies in this review (n = 3) used the biomonitoring approach. Furthermore, variation within the outcome measures make it difficult to compare between several original studies (indirectness assessed in Table 2). It is more difficult to link exposure biomarkers to chronic health outcomes. Nonetheless, questionnaires or sampling information are required to interpret biomonitoring data. A study with direct (biomonitoring) and indirect exposure assessment methods will most likely reduce exposure misclassification. Overall, molecular epidemiological studies should include both biomonitoring for quantification of present exposures and questionnaires to characterize past exposures. Some questionnaires should be added to assess the overall risk associated with PPP practices. For example, to build a combination of exposure determinants, such as IL and CEI. Correlation between the IL and CEI intensity scores and urinary biomarker concentrations were investigated in several studies leading to divergent results [13,49,50,51,52]. These findings lead to the similar conclusion than the meta-analysis that several exposure determinants are required to evaluate occupational exposures. Some findings highlighted missing information in the algorithm, which could explain possible misclassifications such as the PPP formula (i.e., powder, granular), the use of adjuvant, or the condition of spaying (i.e., orchard) [13,51,52].
The hazard of a PPP active ingredient is defined as the inherent property that can cause a health effect [53]. Few studies considered specific active ingredients of PPPs in their exposure assessment. The ones that did showed low or very low quality of evidence for active ingredient association with any health outcome. All studies included in this systematic review disregarded the chemical properties of the active ingredient (e.g., partitioning coefficient, volatility, etc.) as an exposure determinant. The hypothesis is that quantifying PPP exposure does not depend on the active ingredient in occupational exposure itself but rather on how it is used [54]. Moreover, agricultural workers are exposed to a cocktail of active ingredients repeatedly and over many years [55]. The mixture effects of PPPs on human health are not well-established yet [56]; however, the active ingredient remains an important variable to associate exposure to a health outcome. Furthermore, PPP producers and companies formulate their own mixtures of surfactants and active ingredients. Hence, each formulation has its specific trade name and composition. Trade names were not directly used as an exposure determinant in the included studies. Instead, they used trade names to categorize PPPs under an active ingredient or a type of PPP. However, the potential toxicity of surfactants used in PPP formulations might be higher than the active ingredient itself [57]. A recent systematic review emphasized that the toxicological hazards of the mixtures should not be underestimated [58]. It would be important to compare the toxicity between different PPP formulations including similar active ingredient and similar application practices. It might be challenging in epidemiological studies on chronic exposure due to changes of formulation over time. In summary, the active ingredient property itself is not sufficient to assess the exposure risk.
When selecting relevant exposure determinants, it is important to consider the major routes of exposure. Even though it is known that agricultural workers are mainly exposed via skin, none of the studies considered here included exposure determinants to assess specifically the skin exposure. Furthermore, in vitro studies have shown that skin permeation varies depending on the PPP formulation and the dose [11]. The American Health Study (AHS) included dermal exposure assessed as PPE status (e.g., long sleeves, gloves, and goggles.). The PPE status was assessed with a score in this study. Exposures were assessed using certain criteria and grouped into exposure levels that were given a score (1, 1.4, …). To take into account skin exposure, they used an algorithm to reduce the exposure level scores according to PPE status. However, this algorithm simplifies the use of PPE by applying the same reduction factor to all three tasks assessed (mixing, applying and repairing) [2]. Inadvertent ingestion is another poorly assessed route although it could influence the exposure assessment [59]. For example, agricultural workers may not clean the equipment after using PPPs. One typical example is the contamination of PPPs found on the tractor steering wheel [60,61]. They might not have access to water while working in the field and might have their break eating without washing their hands.
In short, to accurately determine PPP exposure, exposure determinants should include the main route of exposure depending on the task and the active ingredient as well as the formulation.
Finally, the exposure determinant definition should be discussed. According to Burstyn and Teschke [7], exposure determinants are used by occupational hygienists and can be applied in epidemiological investigations. Each exposure determinant should always be carefully described and defined with qualifiers of evaluation. As illustrated in Table 1, frequency of application is a good example of inconsistent definition of an exposure determinant. Some studies collected the number of application event per days/week/season [62,63,64,65], while others reported the date of the first PPP application [66,67]. The lack of standardized frequency measurement makes comparisons difficult. The maximum frequency of application determined by regulation could be used to model frequency. It determines how often a PPP can be sprayed per growing season. However, this assumption is based on the worst-case scenario and does not always reflect reality. From our point of view, the most accurate definition for PPP application frequency is the number of applications per day using the hour units. This includes the total period of time worker may be in contact with the PPPs, namely from the beginning to the end of work, regardless of PPE use and types of tasks.

4.3. Strengths and Limitations

This review was based on three of the largest scientific databases and included various keywords using different synonyms to identify relevant studies. Although 25 exposure determinants were listed in the studies, we only included those that were evaluated in a minimum of two studies. Conducting a systematic review of occupational epidemiology, particularly on pesticides, is challenging due to the inherent limitations of the original studies and the heterogeneity of available evidence. The first limitation was the missing information in the studies: 18% of the articles included in this systematic review were of high risk of bias due to missing detailed description of the study participants or appropriate statistical analysis. Further studies should address these issues by providing the information (i.e., open-source publication). Other limitations are the response and recall bias from self-reported questionnaires, the exposure assessment and/or health outcomes assessment that could lower the quality of a study. Risk of bias assessment and the GRADE tools provide to be useful to assess quality studies and therefore the review overall quality.

4.4. Implications of the Findings

This review emphasized a lack of exposure characterization in epidemiological studies. This leads to difficulties in interpreting or associating PPP exposures to health outcomes. A standardized list of exposure determinants for PPP occupational exposures would be of great interest to harmonize occupational studies as well as to increase relevance in protecting human health. Although many studies with valuable exposure data were available, they were not always used by the study authors. Instead, the participants are grouped into dichotomous groups (exposed vs. nonexposed) for comparison. Studies including groups with different exposure levels required a sufficiently high number of participants to obtain a statistically significant result. Data collected from different studies should be combined to improve PPP exposure characterizations. Open-source publications could facilitate better use of these data especially by creating a global registry framework as suggested for human biomonitoring [48]. Harmonized data collection on exposures and health outcomes are required. With a consensus from the scientific community, exposure determinants pertaining to PPP exposures should be listed, units harmonized, and ranked for relevance. To characterize the occupational PPP exposures appropriate data on intensity, duration, and frequency such as IL and CEI exposure calculation, frequency in hours, and amount of PPP applied, are necessary to scale and quantitatively estimate long-term exposures. Data on PPP formulations should be favored over data on active ingredients, because of the higher toxicity of some adjuvants added to formulations compared to the active ingredient itself [57]. PPP formulations combined with other parameters, such as dosage per surface and surface treated, will give a better estimate for PPP exposure. In future studies, biomonitoring could be an alternative to measure internal dose as it takes into account all sources of exposure (occupational as well as environmental exposures and uptake through food or other contaminants) [68]. Before collecting samples in biomonitoring, knowledge on toxicokinetic processes of the active ingredients should be documented to accurately interpret biomonitoring results (internal dose) and define PPP exposures (external dose). Ideally, biomonitoring should include exposure biomarkers and early-effects markers related to health outcomes. This approach would add to the existing health-based data and improve risk assessments. Finally, to reduce reporting bias, health outcome should be as much as possible diagnosed from a physician instead of extracted from self-reported questionnaire.
Overall, there is a need for a standardized list of exposure determinants for PPP exposures in occupational exposure studies. Cohorts may be used to carry out risk assessments, however, specific uniformed data allowing pooling studies are required. Therefore, we recommend defining a standardized list of exposure determinant to collect as initiated by Dosemeci [2].

5. Conclusions

This review highlighted the high variability in epidemiologic studies (e.g., indirectness) and the importance of collecting PPP exposure determinants. The most frequent exposure determinants are the lifetime exposure days and the intensity-weighted exposure days, and we found a moderate quality of evidence for their protective effects against cancer occurrence. A standardized list should at least include intensity, duration, and frequency of PPP exposure as well as formulations. Biomonitoring should also be added to assess PPP exposure and health outcomes when relevant in epidemiological studies because it takes into account dermal uptake which is the main route of exposure in agricultural workers. This could further support to assess the risk for user health, including agricultural worker, for PPP exposure.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/toxics11070623/s1, PRISMA 2020 Checklist; Text S1: String research keywords; Figure S1: Funnel plot on association between active ingredient and cancer for the evaluation of the publication bias; Figure S2: Funnel plot on association between lifetime exposure days and cancer for the evaluation of the publication bias; Figure S3: Funnel plot on association between Intensity-weighted exposure days and cancer for the evaluation of the publication bias; Table S1: 71 Articles included in the meta-analysis with additional data extracted; Table S2: Overall risk of bias assessment following the Joanna Briggs Institute (JBI) checklist for prevalence studies [16,62,64,65,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135].

Author Contributions

Conceptualization, A.B., C.O. and N.B.H.; methodology, C.O., Y.S. and H.G.; software, C.O. and Y.S.; validation, C.O., Z.M. and A.B.; formal analysis, C.O. and Y.S.; investigation, C.O. and A.B.; data curation, C.O. and A.B.; writing—original draft preparation, C.O.; writing—review and editing, C.O., Y.S., Z.M., A.B. and N.B.H.; visualization, C.O.; supervision, A.B. and N.B.H.; project administration, A.B.; funding acquisition, A.B. and N.B.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the State Secretariat of Economic Affairs (SECO). The sponsor had no role in the designing, collecting, analyzing, and interpreting the data.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created in this study. Data sharing is not applicable to this article. The articles included in the meta-analysis are listed in Table S1.

Acknowledgments

We thank Olivier Sanvido and Michel Hauser who helped improve the discussion.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. WHO Patient Safety Conceptual Framework for the International Classification for Patient Safety Version 1.1: Final Technical Report January 2009; World Health Organization: Geneva, Switzerland, 2010; p. 154. [Google Scholar]
  2. Dosemeci, M.; Alavanja, M.C.R.; Rowland, A.S.; Mage, D.; Zahm, S.H.; Rothman, N.; Lubin, J.H.; Hoppin, J.A.; Sandler, D.P.; Blair, A. A Quantitative Approach for Estimating Exposure to Pesticides in the Agricultural Health Study. Ann. Occup. Hyg. 2002, 46, 245–260. [Google Scholar] [CrossRef] [Green Version]
  3. Mandić-Rajčević, S.; Rubino, F.M.; Colosio, C. Exposure and Risk Profiles: From Field Studies to Typical Exposure and Risk Scenarios. In Exposure and Risk Assessment of Pesticide Use in Agriculture; Elsevier: Amsterdam, The Netherlands, 2021; pp. 199–224. ISBN 978-0-12-812466-6. [Google Scholar]
  4. EFSA, (European Food Safety Authority). Guidance on the assessment of exposure of operators, workers, residents and bystanders in risk assessment for plant protection products|Autorité européenne de sécurité des aliments. Wiley Online Libr. 2014, 12, 3874. [Google Scholar] [CrossRef]
  5. Mostafalou, S.; Abdollahi, M. Pesticides: An Update of Human Exposure and Toxicity. Arch. Toxicol. 2017, 91, 549–599. [Google Scholar] [CrossRef]
  6. EFSA. Peer Review of the Pesticide Risk Assessment of the Potential Endocrine Disrupting Properties of Glyphosate. EFSA J. 2017, 15, e04979. [Google Scholar] [CrossRef]
  7. Burstyn, I.; Teschke, K. Studying the Determinants of Exposure: A Review of Methods. Am. Ind. Hyg. Assoc. J. 1999, 60, 57–72. [Google Scholar] [CrossRef]
  8. Kromhout, H.; Symanski, E.; Rappaport, S.M. A comprehensive evaluation of within- and between-worker components of occupational exposure to chemical agents. Ann. Occup. Hyg. 1993, 37, 253–270. [Google Scholar] [CrossRef] [Green Version]
  9. Petit, P.; Bicout, D.J.; Persoons, R.; Bonneterre, V.; Barbeau, D.; Maître, A. Constructing a Database of Similar Exposure Groups: The Application of the Exporisq-HAP Database from 1995 to 2015. Ann. Work Expo. Health 2017, 61, 440–456. [Google Scholar] [CrossRef]
  10. Berthet, A.; Bouchard, M.; Vernez, D. Toxicokinetics of Captan and Folpet Biomarkers in Dermally Exposed Volunteers: Dermal Kinetics of Captan and Folpet Biomarkers. J. Appl. Toxicol. 2012, 32, 202–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Berthet, A.; Hopf, N.B.; Miles, A.; Spring, P.; Charrière, N.; Garrigou, A.; Baldi, I.; Vernez, D. Human Skin in Vitro Permeation of Bentazon and Isoproturon Formulations with or without Protective Clothing Suit. Arch. Toxicol. 2014, 88, 77–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Zahm, S.H.; Ward, M.H.; Blair, A. Pesticides and Cancer. Occup. Med. 1997, 12, 269–289. [Google Scholar] [PubMed]
  13. Acquavella, J.F.; Alexander, B.H.; Mandel, J.S.; Burns, C.J.; Gustin, C. Exposure Misclassification in Studies of Agricultural Pesticides: Insights from Biomonitoring. Epidemiology 2006, 17, 69–74. [Google Scholar] [CrossRef] [PubMed]
  14. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G. Preferred Reporting Items for Systematic Reviews and Meta-Analyses: The PRISMA Statement. J. Clin. Epidemiol. 2009, 62, 1006–1012. [Google Scholar] [CrossRef] [PubMed]
  15. Ouzzani, M.; Hammady, H.; Fedorowicz, Z.; Elmagarmid, A. Rayyan—A Web and Mobile App for Systematic Reviews. Syst. Rev. 2016, 5, 210. [Google Scholar] [CrossRef] [Green Version]
  16. Andreotti, G.; Beane Freeman, L.E.; Shearer, J.J.; Lerro, C.C.; Koutros, S.; Parks, C.G.; Blair, A.; Lynch, C.F.; Lubin, J.H.; Sandler, D.P.; et al. Occupational Pesticide Use and Risk of Renal Cell Carcinoma in the Agricultural Health Study. Env. Health Perspect. 2020, 128, 067011. [Google Scholar] [CrossRef]
  17. Shor, E.; Roelfs, D.; Vang, Z.M. The “Hispanic Mortality Paradox” Revisited: Meta-Analysis and Meta-Regression of Life-Course Differentials in Latin American and Caribbean Immigrants’ Mortality. Soc. Sci. Med. 2017, 186, 20–33. [Google Scholar] [CrossRef]
  18. Zhang, J.; Yu, K.F. What’s the Relative Risk?: A Method of Correcting the Odds Ratio in Cohort Studies of Common Outcomes. JAMA 1998, 280, 1690. [Google Scholar] [CrossRef] [Green Version]
  19. Thorlund, K.; Imberger, G.; Johnston, B.C.; Walsh, M.; Awad, T.; Thabane, L.; Gluud, C.; Devereaux, P.J.; Wetterslev, J. Evolution of Heterogeneity (I2) Estimates and Their 95% Confidence Intervals in Large Meta-Analyses. PLoS ONE 2012, 7, e39471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Higgins, J.P.T.; Thomas, J.; Chandler, J.; Cumpston, M.; Li, T.; Page, M.J.; Welch, V.A. (Eds.) Cochrane Handbook for Systematic Reviews of Interventions Version 6.2 (Updated February 2021). Cochrane. 2021. Available online: www.training.cochrane.org/handbook (accessed on 21 February 2021).
  21. Sutton, A.; Abrams, K.R.; Jones, D.R.; Sheldon, T.; Song, F. Methods for Meta-Analysis in Medical Research; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2000; p. 317. [Google Scholar]
  22. Munn, Z.; Moola, S.; Lisy, K.; Riitano, D.; Tufanaru, C. Methodological guidance for systematic reviews of observational epidemiological studies reporting prevalence and incidence data. Int. J. Evid. Based Healthc. 2015, 13, 147–153. [Google Scholar] [CrossRef]
  23. Ma, L.-L.; Wang, Y.-Y.; Yang, Z.-H.; Huang, D.; Weng, H.; Zeng, X.-T. Methodological Quality (Risk of Bias) Assessment Tools for Primary and Secondary Medical Studies: What Are They and Which Is Better? Mil. Med. Res. 2020, 7, 7. [Google Scholar] [CrossRef] [Green Version]
  24. Guyatt, G.H.; Oxman, A.D.; Vist, G.E.; Kunz, R.; Falck-Ytter, Y.; Alonso-Coello, P.; Schünemann, H.J. GRADE: An Emerging Consensus on Rating Quality of Evidence and Strength of Recommendations. BMJ 2008, 336, 924–926. [Google Scholar] [CrossRef] [Green Version]
  25. Guyatt, G.H.; Oxman, A.D.; Kunz, R.; Woodcock, J.; Brozek, J.; Helfand, M.; Alonso-Coello, P.; Falck-Ytter, Y.; Jaeschke, R.; Vist, G.; et al. GRADE Guidelines: 8. Rating the Quality of Evidence—Indirectness. J. Clin. Epidemiol. 2011, 64, 1303–1310. [Google Scholar] [CrossRef]
  26. Deeks, J.J.; Higgins, J.P.T.; Altman, D.G. Chapter 10: Analysing Data and Undertaking Meta-Analyses. In Cochrane Handbook for Systematic Reviews of Interventions Version 6.2 (Updated February 2021); Higgins, J.P.T., Thomas, J., Chandler, J., Cumpston, M., Li, T., Page, M.J., Welch, V.A., Eds.; Cochrane: London, UK, 2021. [Google Scholar]
  27. Mahadevan, S.; Sadacharan, D.; Kannan, S.; Suryanarayanan, A. Does Time of Sampling or Food Intake Alter Thyroid Function Test? Indian J. Endocr. Metab. 2017, 21, 369. [Google Scholar] [CrossRef]
  28. Fareed, M.; Pathak, M.K.; Bihari, V.; Kamal, R.; Srivastava, A.K.; Kesavachandran, C.N. Adverse Respiratory Health and Hematological Alterations among Agricultural Workers Occupationally Exposed to Organophosphate Pesticides: A Cross-Sectional Study in North India. PLoS ONE 2013, 8, e69755. [Google Scholar] [CrossRef]
  29. Fieten, K.B.; Kromhout, H.; Heederik, D.; van Wendel de Joode, B. Pesticide Exposure and Respiratory Health of Indigenous Women in Costa Rica. Am. J. Epidemiol. 2009, 169, 1500–1506. [Google Scholar] [CrossRef] [PubMed]
  30. Venkata, R.P.; Rahman, M.F.; Mahboob, M.; Kumari, S.I.; Chinde, S.; Bhanuramya, M.; Dumala, N.; Grover, P. Assessment of Genotoxicity in Female Agricultural Workers Exposed to Pesticides. Biomarkers 2017, 22, 446–454. [Google Scholar] [CrossRef] [PubMed]
  31. Burdorf, A. Identification of Determinants of Exposure: Consequences for Measurement and Control Strategies. Occup. Environ. Med. 2005, 62, 344–350. [Google Scholar] [CrossRef] [Green Version]
  32. Hoppin, J.A.; Umbach, D.M.; London, S.J.; Lynch, C.F.; Alavanja, M.C.R.; Sandler, D.P. Pesticides and Adult Respiratory Outcomes in the Agricultural Health Study. Ann. N. Y. Acad. Sci. 2006, 1076, 343–354. [Google Scholar] [CrossRef]
  33. Carles, C.; Bouvier, G.; Lebailly, P.; Baldi, I. Use of Job-Exposure Matrices to Estimate Occupational Exposure to Pesticides: A Review. J. Expo. Sci. Environ. Epidemiol. 2017, 27, 125–140. [Google Scholar] [CrossRef] [PubMed]
  34. Flegal, K.M.; Brownie, C.; Haas, J. The effects of exposure misclassification on estimates of relative risk. Am. J. Epidemiol. 1986, 123, 736–751. [Google Scholar] [CrossRef]
  35. Ohlander, J.; Fuhrimann, S.; Basinas, I.; Cherrie, J.W.; Galea, K.S.; Povey, A.C.; van Tongeren, M.; Harding, A.-H.; Jones, K.; Vermeulen, R.; et al. Systematic Review of Methods Used to Assess Exposure to Pesticides in Occupational Epidemiology Studies, 1993–2017. Occup. Environ. Med. 2020, 77, 357–367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Bureau, M.; Béziat, B.; Duporté, G.; Bouchart, V.; Lecluse, Y.; Barron, E.; Garrigou, A.; Dévier, M.-H.; Budzinski, H.; Lebailly, P.; et al. Pesticide Exposure of Workers in Apple Growing in France. Int. Arch. Occup. Environ. Health 2021, 95, 811–823. [Google Scholar] [CrossRef]
  37. Bouyer, J.; Hemon, D. Studying the Performance of a Job Exposure Matrix. Int. J. Epidemiol. 1993, 22, S65–S71. [Google Scholar] [CrossRef] [PubMed]
  38. De Matteis, S.; Consonni, D. Methods of Exposure Assessment to Pesticides in Agriculture in Epidemiological Studies. In Exposure and Risk Assessment of Pesticide Use in Agriculture; Elsevier: Amsterdam, The Netherlands, 2021; pp. 165–172. ISBN 978-0-12-812466-6. [Google Scholar]
  39. Demetriou, C.; Ozer, B.U.; Essau, C.A. Self-Report Questionnaires. In The Encyclopedia of Clinical Psychology; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2015; pp. 1–6. ISBN 978-1-118-62539-2. [Google Scholar]
  40. Brouwer, M.; Schinasi, L.; Beane Freeman, L.E.; Baldi, I.; Lebailly, P.; Ferro, G.; Nordby, K.-C.; Schüz, J.; Leon, M.E.; Kromhout, H. Assessment of Occupational Exposure to Pesticides in a Pooled Analysis of Agricultural Cohorts within the AGRICOH Consortium. Occup. Environ. Med. 2016, 73, 359–367. [Google Scholar] [CrossRef]
  41. Tsatsakis, A.M.; Alegakis, A.; Katsikantami, I.; Rakitskii, V. Biological Monitoring in Agriculture. In Exposure and Risk Assessment of Pesticide Use in Agriculture; Elsevier: Amsterdam, The Netherlands, 2021; pp. 103–117. ISBN 978-0-12-812466-6. [Google Scholar]
  42. Barr, D.B.; Thomas, K.; Curwin, B.; Landsittel, D.; Raymer, J.; Lu, C.; Donnelly, K.C.; Acquavella, J. Biomonitoring of Exposure in Farmworker Studies. Environ. Health Perspect. 2006, 114, 936–942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Goumenou, M.; Renieri, E.A.; Petrakis, D.; Nathanail, A.V.; Kokaraki, V.; Tsatsakis, A. Methods for Environmental Monitoring of Pesticide Exposure. In Exposure and Risk Assessment of Pesticide Use in Agriculture; Elsevier: Amsterdam, The Netherlands, 2021; pp. 347–387. ISBN 978-0-12-812466-6. [Google Scholar]
  44. Mandić-Rajčević, S.; Rubino, F.M.; Colosio, C. Establishing Health-Based Biological Exposure Limits for Pesticides: A Proof of Principle Study Using Mancozeb. Regul. Toxicol. Pharmacol. 2020, 115, 104689. [Google Scholar] [CrossRef]
  45. Colosio, C.; Rubino, F.M.; Alegakis, A.; Ariano, E.; Brambilla, G.; Mandic-Rajcevic, S.; Metruccio, F.; Minoia, C.; Moretto, A.; Somaruga, C.; et al. Integration of Biological Monitoring, Environmental Monitoring and Computational Modelling into the Interpretation of Pesticide Exposure Data: Introduction to a Proposed Approach. Toxicol. Lett. 2012, 213, 49–56. [Google Scholar] [CrossRef] [PubMed]
  46. Fustinoni, S.; Campo, L.; Colosio, C.; Birindelli, S.; Foà, V. Application of Gas Chromatography-Mass Spectrometry for the Determination of Urinary Ethylenethiourea in Humans. J. Chromatogr. B 2005, 814, 251–258. [Google Scholar] [CrossRef] [PubMed]
  47. Viegas, S.; Zare Jeddi, M.; Hopf, N.B.; Bessems, J.; Palmen, N.; Galea, K.S.; Jones, K.; Kujath, P.; Duca, R.-C.; Verhagen, H.; et al. Biomonitoring as an Underused Exposure Assessment Tool in Occupational Safety and Health Context—Challenges and Way Forward. IJERPH 2020, 17, 5884. [Google Scholar] [CrossRef]
  48. Zare Jeddi, M.; Hopf, N.B.; Louro, H.; Viegas, S.; Galea, K.S.; Pasanen-Kase, R.; Santonen, T.; Mustieles, V.; Fernandez, M.F.; Verhagen, H.; et al. Developing Human Biomonitoring as a 21st Century Toolbox within the European Exposure Science Strategy 2020–2030. Environ. Int. 2022, 168, 107476. [Google Scholar] [CrossRef]
  49. Blair, A.; Thomas, K.; Coble, J.; Sandler, D.P.; Hines, C.J.; Lynch, C.F.; Knott, C.; Purdue, M.P.; Zahm, S.H.; Alavanja, M.C.R.; et al. Impact of Pesticide Exposure Misclassification on Estimates of Relative Risks in the Agricultural Health Study. Occup. Environ. Med. 2011, 68, 537–541. [Google Scholar] [CrossRef] [Green Version]
  50. Coble, J.; Arbuckle, T.; Lee, W.; Alavanja, M.; Dosemeci, M. The Validation of a Pesticide Exposure Algorithm Using Biological Monitoring Results. J. Occup. Environ. Hyg 2005, 2, 194–201. [Google Scholar] [CrossRef] [PubMed]
  51. Hines, C.J.; Deddens, J.A.; Jaycox, L.B.; Andrews, R.N.; Striley, C.A.F.; Alavanja, M.C.R. Captan Exposure and Evaluation of a Pesticide Exposure Algorithm among Orchard Pesticide Applicators in the Agricultural Health Study. Ann. Occup. Hyg. 2008, 52, 153–166. [Google Scholar] [CrossRef]
  52. Thomas, K.W.; Dosemeci, M.; Coble, J.B.; Hoppin, J.A.; Sheldon, L.S.; Chapa, G.; Croghan, C.W.; Jones, P.A.; Knott, C.E.; Lynch, C.F.; et al. Assessment of a Pesticide Exposure Intensity Algorithm in the Agricultural Health Study. J. Expo. Sci. Environ. Epidemiol. 2010, 20, 559–569. [Google Scholar] [CrossRef]
  53. IPCS. Risk Assessment Terminology; Inter-Organization Programme for the Sound Management of Chemicals, International Program on Chemical Safety, Eds.; Harmonization Project Document; OMS: Geneve, Switzerland, 2004; ISBN 978-92-4-156267-6.
  54. Metruccio, F.; Tosti, L.; Moretto, A. Models Used in the World (European Union, United States, Other Countries). In Exposure and Risk Assessment of Pesticide Use in Agriculture; Elsevier: Amsterdam, The Netherlands, 2021; pp. 25–67. ISBN 978-0-12-812466-6. [Google Scholar]
  55. Baldi, I.; Carles, C.; Blanc-Lapierre, A.; Fabbro-Peray, P.; Druet-Cabanac, M.; Boutet-Robinet, E.; Soulat, J.-M.; Bouvier, G.; Lebailly, P.; The PESTIMAT Group; et al. A French Crop-Exposure Matrix for Use in Epidemiological Studies on Pesticides: PESTIMAT. J. Expo. Sci. Environ. Epidemiol. 2017, 27, 56–63. [Google Scholar] [CrossRef] [PubMed]
  56. De-Assis, M.P.; Barcella, R.C.; Padilha, J.C.; Pohl, H.H.; Krug, S.B.F. Health Problems in Agricultural Workers Occupationally Exposed to Pesticides. Rev. Bras. Med. Trab. 2020, 18, 352–363. [Google Scholar] [CrossRef]
  57. Mesnage, R.; Antoniou, M.N. Ignoring Adjuvant Toxicity Falsifies the Safety Profile of Commercial Pesticides. Front. Public Health 2018, 5, 361. [Google Scholar] [CrossRef] [Green Version]
  58. Nagy, K.; Duca, R.C.; Lovas, S.; Creta, M.; Scheepers, P.T.J.; Godderis, L.; Ádám, B. Systematic Review of Comparative Studies Assessing the Toxicity of Pesticide Active Ingredients and Their Product Formulations. Environ. Res. 2020, 181, 108926. [Google Scholar] [CrossRef]
  59. Côté, J.; Bouchard, M. Dose Reconstruction in Workers Exposed to Two Major Pyrethroid Pesticides and Determination of Biological Reference Values Using a Toxicokinetic Model. J. Expo. Sci. Environ. Epidemiol. 2018, 28, 599–614. [Google Scholar] [CrossRef] [PubMed]
  60. Cherrie, J.W.; Semple, S.; Christopher, Y.; Saleem, A.; Hughson, G.W.; Philips, A. How Important Is Inadvertent Ingestion of Hazardous Substances at Work? Ann. Occup. Hyg. 2006, 50, 693–704. [Google Scholar] [CrossRef] [Green Version]
  61. Tudi, M.; Li, H.; Li, H.; Wang, L.; Lyu, J.; Yang, L.; Tong, S.; Yu, Q.J.; Ruan, H.D.; Atabila, A.; et al. Exposure Routes and Health Risks Associated with Pesticide Application. Toxics 2022, 10, 335. [Google Scholar] [CrossRef]
  62. Boulanger, M.; Tual, S.; Lemarchand, C.; Guizard, A.-V.; Velten, M.; Marcotullio, E.; Baldi, I.; Clin, B.; Lebailly, P. Agricultural Exposure and Risk of Bladder Cancer in the AGRIculture and CANcer Cohort. Int. Arch. Occup. Environ. Health 2017, 90, 169–178. [Google Scholar] [CrossRef] [PubMed]
  63. Hu, R.; Huang, X.; Huang, J.; Li, Y.; Zhang, C.; Yin, Y.; Chen, Z.; Jin, Y.; Cai, J.; Cui, F. Long- and Short-Term Health Effects of Pesticide Exposure: A Cohort Study from China. PLoS ONE 2015, 10, e0128766. [Google Scholar] [CrossRef] [PubMed]
  64. Piel, C.; Pouchieu, C.; Tual, S.; Migault, L.; Lemarchand, C.; Carles, C.; Boulanger, M.; Gruber, A.; Rondeau, V.; Marcotullio, E.; et al. Central Nervous System Tumors and Agricultural Exposures in the Prospective Cohort AGRICAN: CNS Tumors and Agricultural Exposures in AGRICAN. Int. J. Cancer 2017, 141, 1771–1782. [Google Scholar] [CrossRef]
  65. Pouchieu, C.; Piel, C.; Carles, C.; Gruber, A.; Helmer, C.; Tual, S.; Marcotullio, E.; Lebailly, P.; Baldi, I. Pesticide Use in Agriculture and Parkinson’s Disease in the AGRICAN Cohort Study. Int. J. Epidemiol. 2018, 47, 299–310. [Google Scholar] [CrossRef] [PubMed]
  66. Baldi, I.; Gruber, A.; Rondeau, V.; Lebailly, P.; Brochard, P.; Fabrigoule, C. Neurobehavioral Effects of Long-Term Exposure to Pesticides: Results from the 4-Year Follow-up of the PHYTONER Study. Occup. Environ. Med. 2011, 68, 108–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Cremonese, C.; Piccoli, C.; Pasqualotto, F.; Clapauch, R.; Koifman, R.J.; Koifman, S.; Freire, C. Occupational Exposure to Pesticides, Reproductive Hormone Levels and Sperm Quality in Young Brazilian Men. Reprod. Toxicol. 2017, 67, 174–185. [Google Scholar] [CrossRef]
  68. Zare Jeddi, M.; Virgolino, A.; Fantke, P.; Hopf, N.B.; Galea, K.S.; Remy, S.; Viegas, S.; Mustieles, V.; Fernandez, M.F.; von Goetz, N.; et al. A Human Biomonitoring (HBM) Global Registry Framework: Further Advancement of HBM Research Following the FAIR Principles. Int. J. Hyg. Environ. Health 2021, 238, 113826. [Google Scholar] [CrossRef]
  69. Alavanja, M.C.R.; Samanic, C.; Dosemeci, M.; Lubin, J.; Tarone, R.; Lynch, C.F.; Knott, C.; Thomas, K.; Hoppin, J.A.; Barker, J.; et al. Use of agricultural pesticides and prostate cancer risk in the Agricultural Health Study cohort. Am. J. Epidemiol. 2003, 157, 800–814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Alavanja, M.C.R.; Dosemeci, M.; Samanic, C.; Lubin, J.H.; Lynch, C.F.; Knott, C.; Barker, J.; Hoppin, J.A.; Sandler, D.P.; Coble, J.; et al. Pesticides and Lung Cancer Risk in the Agricultural Health Study Cohort. Am. J. Epidemiol. 2004, 160, 876–885. [Google Scholar] [CrossRef]
  71. Alavanja, M.C.R.; Hofmann, J.N.; Lynch, C.F.; Hines, C.J.; Barry, K.H.; Barker, J.; Buckman, D.W.; Thomas, K.; Sandler, D.P.; Hoppin, J.A.; et al. Non-hodgkin lymphoma risk and insecticide, fungicide and fumigant use in the agricultural health study. PLoS ONE 2014, 9, e109332. [Google Scholar] [CrossRef]
  72. Andreotti, G.; Freeman, L.E.; Hou, L.; Coble, J.; Rusiecki, J.; Hoppin, J.A.; Silverman, D.T.; Alavanja, M.C.R. Agricultural pesticide use and pancreatic cancer risk in the Agricultural Health Study Cohort. Int. J. Cancer 2009, 124, 2495–2500. [Google Scholar] [CrossRef] [Green Version]
  73. Andreotti, G.; Koutros, S.; Hofmann, J.N.; Sandler, D.P.; Lubin, J.H.; Lynch, C.F.; Lerro, C.C.; De Roos, A.J.; Parks, C.G.; Alavanja, M.C.R.; et al. Glyphosate Use and Cancer Incidence in the Agricultural Health Study. J. Natl. Cancer Inst. 2018, 110, 509–516. [Google Scholar] [CrossRef] [Green Version]
  74. Band, P.R.; Abanto, Z.; Bert, J.; Lang, B.; Fang, R.; Gallagher, R.P.; Le, N.D. Prostate cancer risk and exposure to pesticides in British Columbia farmers. Prostate 2011, 71, 168–183. [Google Scholar] [CrossRef] [PubMed]
  75. Barry, K.H.; Koutros, S.; Lubin, J.H.; Coble, J.B.; Barone-Adesi, F.; Beane Freeman, L.E.; Sandler, D.P.; Hoppin, J.A.; Ma, X.; Zheng, T.; et al. Methyl bromide exposure and cancer risk in the Agricultural Health Study. Cancer Causes Control. 2012, 23, 807–818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Beane Freeman, L.E.; Bonner, M.R.; Blair, A.; Hoppin, J.A.; Sandler, D.P.; Lubin, J.H.; Dosemeci, M.; Lynch, C.F.; Knott, C.; Alavanja, M.C.R. Cancer incidence among male pesticide applicators in the Agricultural Health Study cohort exposed to diazinon. Am. J. Epidemiol. 2005, 162, 1070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Bonner, M.R.; Coble, J.; Blair, A.; Beane Freeman, L.E.; Hoppin, J.A.; Sandler, D.P.; Alavanja, M.C.R. Malathion exposure and the incidence of cancer in the agricultural health study. Am. J. Epidemiol. 2007, 166, 1023–1034. [Google Scholar] [CrossRef] [PubMed]
  78. Bonner, M.R.; Williams, B.A.; Rusiecki, J.A.; Blair, A.; Beane Freeman, L.E.; Hoppin, J.A.; Dosemeci, M.; Lubin, J.; Sandler, D.P.; Alavanja, M.C.R. Occupational exposure to terbufos and the incidence of cancer in the Agricultural Health Study. Cancer Causes Control. 2010, 21, 871–877. [Google Scholar] [CrossRef]
  79. Bonner, M.R.; Freeman, L.E.; Hoppin, J.A.; Koutros, S.; Sandler, D.P.; Lynch, C.F.; Hines, C.J.; Thomas, K.; Blair, A.; Alavanja, M.C.R. Occupational Exposure to Pesticides and the Incidence of Lung Cancer in the Agricultural Health Study. Environ. Health Perspect. 2017, 125, 544–551. [Google Scholar] [CrossRef] [Green Version]
  80. Boulanger, M.; Tual, S.; Lemarchand, C.; Guizard, A.V.; Delafosse, P.; Marcotullio, E.; Pons, R.; Piel, C.; Pouchieu, C.; Baldi, I.; et al. Lung cancer risk and occupational exposures in crop farming: Results from the AGRIculture and CANcer (AGRICAN) cohort. Occup. Environ. Med. 2018, 75, 776–785. [Google Scholar] [CrossRef]
  81. Chiu, B.C.; Dave, B.J.; Blair, A.; Gapstur, S.M.; Zahm, S.H.; Weisenburger, D.D. Agricultural pesticide use and risk of t(14;18)-defined subtypes of non-Hodgkin lymphoma. Blood 2006, 108, 1363–1369. [Google Scholar] [CrossRef] [Green Version]
  82. Christensen, C.H.; Platz, E.A.; Andreotti, G.; Blair, A.; Hoppin, J.A.; Koutros, S.; Lynch, C.F.; Sandler, D.P.; Alavanja, M.C.R. Coumaphos exposure and incident cancer among male participants in the Agricultural Health Study (AHS). Environ. Health Perspect. 2010, 118, 92–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. De Roos, A.J.; Blair, A.; Rusiecki, J.A.; Hoppin, J.A.; Svec, M.; Dosemeci, M.; Sandler, D.P.; Alavanja, M.C.R. Cancer incidence among glyphosate-exposed pesticide applicators in the Agricultural Health Study. Environ. Health Perspect. 2005, 113, 49–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Delancey, J.O.; Alavanja, M.C.; Coble, J.; Blair, A.; Hoppin, J.A.; Austin, H.D.; Beane Freeman, L.E. Occupational exposure to metribuzin and the incidence of cancer in the Agricultural Health Study. Ann. Epidemiol. 2009, 19, 388–395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Dennis, L.K.; Lynch, C.F.; Sandler, D.P.; Alavanja, M.C.R. Pesticide use and cutaneous melanoma in pesticide applicators in the agricultural heath study. Environ. Health Perspect. 2010, 118, 812–817. [Google Scholar] [CrossRef] [Green Version]
  86. Engel, L.S.; Hill, D.A.; Hoppin, J.A.; Lubin, J.H.; Lynch, C.F.; Pierce, J.; Samanic, C.; Sandler, D.P.; Blair, A.; Alavanja, M.C.R. Pesticide use and breast cancer risk among farmers’ wives in the agricultural health study. Am. J. Epidemiol. 2005, 161, 121–135. [Google Scholar] [CrossRef]
  87. Goldner, W.S.; Sandler, D.P.; Yu, F.; Shostrom, V.; Hoppin, J.A.; Kamel, F.; LeVan, T.D. Hypothyroidism and pesticide use among male private pesticide applicators in the agricultural health study. J. Occup. Environ. Med. 2013, 55, 1171. [Google Scholar] [CrossRef] [Green Version]
  88. Greenburg, D.L.; Rusiecki, J.; Koutros, S.; Dosemeci, M.; Patel, R.; Hines, C.J.; Hoppin, J.A.; Alavanja, M.C. Cancer incidence among pesticide applicators exposed to captan in the Agricultural Health Study. Cancer Causes Control. 2008, 19, 1401. [Google Scholar] [CrossRef]
  89. Hofmann, J.N.; Beane Freeman, L.E.; Murata, K.; Andreotti, G.; Shearer, J.J.; Thoren, K.; Ramanathan, L.; Parks, C.G.; Koutros, S.; Lerro, C.C.; et al. Lifetime Pesticide Use and Monoclonal Gammopathy of Undetermined Significance in a Prospective Cohort of Male Farmers. Environ. Health Perspect. 2021, 129, 17003. [Google Scholar] [CrossRef]
  90. Hou, L.; Lee, W.J.; Rusiecki, J.; Hoppin, J.A.; Blair, A.; Bonner, M.R.; Lubin, J.H.; Samanic, C.; Sandler, D.P.; Dosemeci, M. Pendimethalin exposure and cancer incidence among pesticide applicators. Epidemiology 2006, 17, 302–307. [Google Scholar] [CrossRef]
  91. Ji, B.T.; Silverman, D.T.; Stewart, P.A.; Blair, A.; Swanson, G.M.; Baris, D.; Greenberg, R.S.; Hayes, R.B.; Brown, L.M.; Lillemoe, K.D.; et al. Occupational exposure to pesticides and pancreatic cancer. Am. J. Ind. Med. 2001, 39, 92–99. [Google Scholar] [CrossRef]
  92. Jones, R.R.; Barone-Adesi, F.; Koutros, S.; Lerro, C.C.; Blair, A.; Lubin, J.; Heltshe, S.L.; Hoppin, J.A.; Alavanja, M.C.; Beane Freeman, L.E. Incidence of solid tumours among pesticide applicators exposed to the organophosphate insecticide diazinon in the Agricultural Health Study: An updated analysis. Occup. Environ. Med. 2015, 72, 496–503. [Google Scholar] [CrossRef] [PubMed]
  93. Kamel, F.; Engel, L.S.; Gladen, B.C.; Hoppin, J.A.; Alavanja, M.C.R.; Sandler, D.P. Neurologic Symptoms in Licensed Private Pesticide Applicators in the Agricultural Health Study. Environ. Health Perspect. 2005, 113, 877–882. [Google Scholar] [CrossRef] [Green Version]
  94. Kang, D.; Park, S.K.; Beane-Freeman, L.; Lynch, C.F.; Knott, C.E.; Sandler, D.P.; Hoppin, J.A.; Dosemeci, M.; Coble, J.; Lubin, J.; et al. Cancer incidence among pesticide applicators exposed to trifluralin in the Agricultural Health Study. Environ. Res. 2008, 107, 271–276. [Google Scholar] [CrossRef]
  95. Koutros, S.; Mahajan, R.; Zheng, T.; Hoppin, J.A.; Ma, X.; Lynch, C.F.; Blair, A.; Alavanja, M.C.R. Dichlorvos exposure and human cancer risk: Results from the Agricultural Health Study. Cancer Causes Control. 2008, 19, 59–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Koutros, S.; Lynch, C.F.; Ma, X.; Lee, W.J.; Hoppin, J.A.; Christensen, C.H.; Andreotti, G.; Freeman, L.B.; Rusiecki, J.A.; Hou, L.; et al. Heterocyclic aromatic amine pesticide use and human cancer risk: Results from the U.S. Agricultural Health Study. Int. J. Cancer 2009, 124, 1206–1212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Koutros, S.; Freeman, L.E.B.; Lubin, J.H.; Heltshe, S.L.; Andreotti, G.; Barry, K.H.; DellaValle, C.T.; Hoppin, J.A.; Sandler, D.P.; Lynch, C.F.; et al. Risk of Total and Aggressive Prostate Cancer and Pesticide Use in the Agricultural Health Study. Am. J. Epidemiology 2013, 177, 59–74. [Google Scholar] [CrossRef]
  98. Koutros, S.; Silverman, D.T.; Alavanja, M.C.; Andreotti, G.; Lerro, C.C.; Heltshe, S.; Lynch, C.F.; Sandler, D.P.; Blair, A.; Freeman, L.E.B. Occupational exposure to pesticides and bladder cancer risk. Int. J. Epidemiol. 2016, 45, 792–805. [Google Scholar] [CrossRef] [Green Version]
  99. Lee, W.J.; Hoppin, J.A.; Blair, A.; Lubin, J.H.; Dosemeci, M.; Sandler, D.P.; Alavanja, M.C.R. Cancer Incidence among Pesticide Applicators Exposed to Alachlor in the Agricultural Health Study. Am. J. Epidemiology 2004, 159, 373–380. [Google Scholar] [CrossRef]
  100. Lee, W.J.; Blair, A.; Hoppin, J.A.; Lubin, J.H.; Rusiecki, J.A.; Sandler, D.P.; Dosemeci, M.; Alavanja, M.C.R. Cancer Incidence Among Pesticide Applicators Exposed to Chlorpyrifos in the Agricultural Health Study. Gynecol. Oncol. 2004, 96, 1781–1789. [Google Scholar] [CrossRef] [Green Version]
  101. Lee, W.J.; Sandler, D.P.; Blair, A.; Samanic, C.; Cross, A.J.; Alavanja, M.C. Pesticide use and colorectal cancer risk in the agricultural health study. Int. J. Cancer 2007, 121, 339–346. [Google Scholar] [CrossRef] [Green Version]
  102. LeMarchand, C.; Tual, S.; Boulanger, M.; Levêque-Morlais, N.; Perrier, S.; Clin, B.; Guizard, A.-V.; Velten, M.; Rigaud, E.; Baldi, I.; et al. Prostate cancer risk among French farmers in the AGRICAN cohort. Scand. J. Work. Environ. Health 2016, 42, 144–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Lerro, C.C.; Koutros, S.; Andreotti, G.; Hines, C.J.; Blair, A.; Lubin, J.; Ma, X.; Zhang, Y.; Freeman, L.E.B. Use of acetochlor and cancer incidence in the Agricultural Health Study. Int. J. Cancer 2015, 137, 1167–1175. [Google Scholar] [CrossRef] [Green Version]
  104. Lerro, C.C.; Hofmann, J.N.; Andreotti, G.; Koutros, S.; Parks, C.G.; Blair, A.; Albert, P.S.; Lubin, J.H.; Sandler, D.P.; Freeman, L.E.B. Dicamba use and cancer incidence in the agricultural health study: An updated analysis. Leuk. Res. 2020, 49, 1326–1337. [Google Scholar] [CrossRef] [PubMed]
  105. Lerro, C.C.; Freeman, L.E.B.; DellaValle, C.T.; Andreotti, G.; Hofmann, J.N.; Koutros, S.; Parks, C.G.; Shrestha, S.; Alavanja, M.C.; Blair, A.; et al. Pesticide exposure and incident thyroid cancer among male pesticide applicators in agricultural health study. Environ. Int. 2021, 146, 106187. [Google Scholar] [CrossRef] [PubMed]
  106. Lerro, C.C.; Freeman, L.E.B.; DellaValle, C.T.; Kibriya, M.G.; Aschebrook-Kilfoy, B.; Jasmine, F.; Koutros, S.; Parks, C.G.; Sandler, D.P.; Alavanja, M.C.R.; et al. Occupational pesticide exposure and subclinical hypothyroidism among male pesticide applicators. Occup. Environ. Med. 2018, 75, 79–89. [Google Scholar] [CrossRef]
  107. Lynch, S.M.; Rusiecki, J.A.; Blair, A.; Dosemeci, M.; Lubin, J.; Sandler, D.; Hoppin, J.A.; Lynch, C.F.; Alavanja, M.C. Cancer Incidence among Pesticide Applicators Exposed to Cyanazine in theAgricultural Health Study. Environ. Health Perspect. 2006, 114, 1248–1252. [Google Scholar] [CrossRef] [Green Version]
  108. Lynch, S.M.; Mahajan, R.; Freeman, L.E.B.; Hoppin, J.A.; Alavanja, M.C. Cancer incidence among pesticide applicators exposed to butylate in the Agricultural Health Study (AHS). Environ. Res. 2009, 109, 860–868. [Google Scholar] [CrossRef] [Green Version]
  109. Mahajan, R.; Bonner, M.R.; Hoppin, J.A.; Alavanja, M.C. Phorate Exposure and Incidence of Cancer in the Agricultural Health Study. Environ. Health Perspect. 2006, 114, 1205–1209. [Google Scholar] [CrossRef] [Green Version]
  110. Mahajan, R.; Blair, A.; Lynch, C.F.; Schroeder, P.; Hoppin, J.A.; Sandler, D.P.; Alavanja, M.C. Fonofos Exposure and Cancer Incidence in the Agricultural Health Study. Environ. Health Perspect. 2006, 114, 1838–1842. [Google Scholar] [CrossRef] [Green Version]
  111. Mahajan, R.; Blair, A.; Coble, J.; Lynch, C.F.; Hoppin, J.A.; Sandler, D.P.; Alavanja, M.C. Carbaryl exposure and incident cancer in the Agricultural Health Study. Int. J. Cancer 2007, 121, 1799–1805. [Google Scholar] [CrossRef]
  112. Mills, P.K.; Yang, R. Prostate Cancer Risk in California Farm Workers. J. Occup. Environ. Med. 2003, 45, 249–258. [Google Scholar] [CrossRef] [PubMed]
  113. Mills, P.K.; Yang, R.; Riordan, D. Lymphohematopoietic Cancers in the United Farm Workers of America (UFW), 1988–2001. Cancer Causes Control. 2005, 16, 823–830. [Google Scholar] [CrossRef] [PubMed]
  114. Montgomery, M.P.; Kamel, F.; Saldana, T.M.; Alavanja, M.C.R.; Sandler, D.P. Incident Diabetes and Pesticide Exposure among Licensed Pesticide Applicators: Agricultural Health Study, 1993-2003. Am. J. Epidemiology 2008, 167, 1235–1246. [Google Scholar] [CrossRef] [PubMed]
  115. Mozzachio, A.M.; Rusiecki, J.A.; Hoppin, J.A.; Mahajan, R.; Patel, R.; Beane-Freeman, L.; Alavanja, M.C. Chlorothalonil exposure and cancer incidence among pesticide applicator participants in the agricultural health study. Environ. Res. 2008, 108, 400–403. [Google Scholar] [CrossRef] [Green Version]
  116. Narayan, S.; Liew, Z.; Bronstein, J.M.; Ritz, B. Occupational pesticide use and Parkinson’s disease in the Parkinson Environment Gene (PEG) study. Environ. Int. 2017, 107, 266–273. [Google Scholar] [CrossRef]
  117. Pardo, L.A.; Freeman, L.E.B.; Lerro, C.C.; Andreotti, G.; Hofmann, J.N.; Parks, C.G.; Sandler, D.P.; Lubin, J.H.; Blair, A.; Koutros, S. Pesticide exposure and risk of aggressive prostate cancer among private pesticide applicators. Environ. Health 2020, 19, 1–12. [Google Scholar] [CrossRef] [Green Version]
  118. Park, S.K.; Kang, D.; Beane-Freeman, L.; Blair, A.; Hoppin, J.A.; Sandler, D.P.; Lynch, C.F.; Knott, C.; Gwak, J.; Alavanja, M. Cancer incidence among paraquat exposed applicators in the agricultural health study: Prospective cohort study. Int. J. Occup. Environ. Health 2009, 15, 274–281. [Google Scholar] [CrossRef]
  119. Park, S.; Kim, S.-K.; Kim, J.-Y.; Lee, K.; Choi, J.R.; Chang, S.-J.; Chung, C.H.; Park, K.-S.; Oh, S.-S.; Koh, S.-B. Exposure to pesticides and the prevalence of diabetes in a rural population in Korea. Neurotoxicology 2019, 70, 12–18. [Google Scholar] [CrossRef]
  120. Piel, C.; Pouchieu, C.; Migault, L.; Béziat, B.; Boulanger, M.; Bureau, M.; Carles, C.; Grüber, A.; Lecluse, Y.; Rondeau, V.; et al. Increased risk of central nervous system tumours with carbamate insecticide use in the prospective cohort AGRICAN. Leuk. Res. 2019, 48, 512–526. [Google Scholar] [CrossRef]
  121. Piel, C.; Pouchieu, C.; Carles, C.; Béziat, B.; Boulanger, M.; Bureau, M.; Busson, A.; Grüber, A.; Lecluse, Y.; Migault, L.; et al. Agricultural exposures to carbamate herbicides and fungicides and central nervous system tumour incidence in the cohort AGRICAN. Environ. Int. 2019, 130, 104876. [Google Scholar] [CrossRef]
  122. Purdue, M.P.; Hoppin, J.A.; Blair, A.; Dosemeci, M.; Alavanja, M.C. Occupational exposure to organochlorine insecticides and cancer incidence in the Agricultural Health Study. Int. J. Cancer 2006, 120, 642–649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Renier, M.; Busson, A.; Boulanger, M.; Piel, C.; Pons, R.; Tual, S.; Amadéo, B.; Meryet-Figuiere, M.; Marcotullio, E.; Clin, B.; et al. Agricultural exposure and risk of soft tissue sarcomas and gastrointestinal stromal sarcoma in the AGRIculture and CANcer (AGRICAN) cohort. Int. J. Cancer 2022, 150, 1792–1803. [Google Scholar] [CrossRef] [PubMed]
  124. Rusiecki, J.A.; De Roos, A.; Lee, W.J.; Dosemeci, M.; Lubin, J.H.; Hoppin, J.A.; Blair, A.; Alavanja, M.C.R. Cancer Incidence Among Pesticide Applicators Exposed to Atrazine in the Agricultural Health Study. Gynecol. Oncol. 2004, 96, 1375–1382. [Google Scholar] [CrossRef] [PubMed]
  125. Rusiecki, J.A.; Hou, L.; Lee, W.J.; Blair, A.; Dosemeci, M.; Lubin, J.H.; Bonner, M.; Samanic, C.; Hoppin, J.A.; Sandler, D.P.; et al. Cancer incidence among pesticide applicators exposed to metolachlor in the Agricultural Health Study. Int. J. Cancer 2006, 118, 3118–3123. [Google Scholar] [CrossRef]
  126. Rusiecki, J.A.; Patel, R.; Koutros, S.; Beane-Freeman, L.; Landgren, O.; Bonner, M.R.; Coble, J.; Lubin, J.; Blair, A.; Hoppin, J.A.; et al. Cancer Incidence among Pesticide Applicators Exposed to Permethrin in the Agricultural Health Study. Environ. Health Perspect. 2009, 117, 581–586. [Google Scholar] [CrossRef] [Green Version]
  127. Samanic, C.; Rusiecki, J.; Dosemeci, M.; Hou, L.; Hoppin, J.A.; Sandler, D.P.; Lubin, J.; Blair, A.; Alavanja, M.C.R. Cancer Incidence among Pesticide Applicators Exposed to Dicamba in the Agricultural Health Study. Environ. Health Perspect. 2006, 114, 1521–1526. [Google Scholar] [CrossRef] [Green Version]
  128. Shrestha, S.; Parks, C.G.; Umbach, D.M.; Richards-Barber, M.; Hofmann, J.N.; Chen, H.; Blair, A.; Freeman, L.E.B.; Sandler, D.P. Pesticide use and incident Parkinson’s disease in a cohort of farmers and their spouses. Environ. Res. 2020, 191, 110186. [Google Scholar] [CrossRef]
  129. Shrestha, S.; Parks, C.G.; Goldner, W.S.; Kamel, F.; Umbach, D.M.; Ward, M.H.; Lerro, C.C.; Koutros, S.; Hofmann, J.N.; Freeman, L.E.B.; et al. Pesticide Use and Incident Hypothyroidism in Pesticide Applicators in the Agricultural Health Study. Environ. Health Perspect. 2018, 126, 97008. [Google Scholar] [CrossRef] [Green Version]
  130. Silver, S.R.; Bertke, S.J.; Hines, C.J.; Alavanja, M.C.R.; Hoppin, J.A.; Lubin, J.H.; Rusiecki, J.A.; Sandler, D.P.; Freeman, L.E.B. Cancer incidence and metolachlor use in the Agricultural Health Study: An update. Int. J. Cancer 2015, 137, 2630–2643. [Google Scholar] [CrossRef] [Green Version]
  131. Starks, S.E.; Hoppin, J.A.; Kamel, F.; Lynch, C.F.; Jones, M.P.; Alavanja, M.C.; Sandler, D.P.; Gerr, F. Peripheral Nervous System Function and Organophosphate Pesticide Use among Licensed Pesticide Applicators in the Agricultural Health Study. Environ. Health Perspect. 2012, 120, 515–520. [Google Scholar] [CrossRef] [Green Version]
  132. Starling, A.P.; Umbach, D.M.; Kamel, F.; Long, S.; Sandler, D.P.; A Hoppin, J. Pesticide use and incident diabetes among wives of farmers in the Agricultural Health Study. Occup. Environ. Med. 2014, 71, 629–635. [Google Scholar] [CrossRef] [PubMed]
  133. Talibov, M.; Tual, S.; Morlais, F.; Meryet-Figuière, M.; Boulanger, M.; Bouvier, V.; Perrier, S.; Clin, B.; Baldi, I.; Lebailly, P.; et al. Colorectal cancer among farmers in the AGRICAN cohort study. Cancer Epidemiology 2022, 78, 102125. [Google Scholar] [CrossRef]
  134. Tual, S.; Busson, A.; Boulanger, M.; Renier, M.; Piel, C.; Pouchieu, C.; Pons, R.; Perrier, S.; Levêque-Morlais, N.; AGRICAN-Group; et al. Occupational exposure to pesticides and multiple myeloma in the AGRICAN cohort. Cancer Causes Control. 2019, 30, 1243–1250. [Google Scholar] [CrossRef] [PubMed]
  135. van Bemmel, D.M.; Visvanathan, K.; Freeman, L.E.B.; Coble, J.; Hoppin, J.A.; Alavanja, M.C.R. S-Ethyl-N,N-dipropylthiocarbamate Exposure and Cancer Incidence among Male Pesticide Applicators in the Agricultural Health Study: A Prospective Cohort. Environ. Health Perspect. 2008, 116, 1541–1546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. PRISMA 2020 flow chart of publication selection.
Figure 1. PRISMA 2020 flow chart of publication selection.
Toxics 11 00623 g001
Table 1. Variable studied and their respective definition, unit of measurement and use in the intensity level (IL) or cumulative exposure intensity (CEI) calculation in the included studies on occupational exposure to PPPs.
Table 1. Variable studied and their respective definition, unit of measurement and use in the intensity level (IL) or cumulative exposure intensity (CEI) calculation in the included studies on occupational exposure to PPPs.
VariableDefinitionMeasureUsed in IL or CEI 1
Socio-professional information
AgeDate of birthYears
GenderMen/Women (M/W) or Male/Female (M/F)M/W or M/F
Job titleEmployment status (e.g., Manager; Workers; Employee) or Self-reported statusTitle
General farming information
CropCategory of cultivated plants (e.g., cereals; fruits; vegetables)Crop type
Cultivated SurfaceLand area used for cropsAcres/Hectare
TaskPiece of work performed within the job (e.g., PPP spraying; harvesting; re-entering)Category of job or task
PPP use
Active ingredientBiological active chemical compoundChemical name
Chemical classGroup of compounds with similar features (i.e., organophosphates, pyrethroids, carbamates, …)Chemical class
Type of PPPTarget pest category of PPP (e.g., fungicide;
herbicide; insecticide)
ClassificationX
PPP licenseObtained date of permit to apply PPPyears
Read instructionOperator read the use instruction provided on the label (e.g., dilution; personal protective equipment; mixing)yes/no
Mixing/loadingOperator prepared the PPP solution and filled the spraying tankyes/noX
Detailed
Application methodSpraying tank type (e.g., backpack sprayer; broadcast from farm vehicle; air sprayer)CategoriesX
Detailed
Tank volumeAmount of PPP solution contained in a tankGallon: liter (3.8:1)
CleaningOperator washed the application materialyes/noDetailed
RepairOperator repaired engineyes/noX
Re-entryOperator entered in field following treatmentyes/no
DurationAmount of time spent on applicationhours or days
FrequencyNumber of years an operator applied PPP
or
Number of days per year an operator applied PPP
or
Number of months an operator applied PPP per cropping season
or
Number of application events per day/week/season
or
Number of application events over the last 12 months
or
Date of first PPP application
year

number

number

number

number

date
Percentage of treated areaPercentage of cultivated surface treated by an
operator
percentage
PPE useUse of PPE while performing PPP tasks
and/or
Use of PPE while cleaning
and/or
Use of PPE after a PPP spill
and/or
Duration of use before changing gloves
yes/no

yes/no

yes/no

hours
X

Detailed

Detailed

Detailed
Personal hygieneOperator washed their hands after PPP applicationyes/noDetailed
PPE: Personal protective equipment; PPP: Plant protection product. 1 Variable used in the calculation of the IL (Intensity Level) or CEI (Cumulative Exposure Index) [2]. “X” means that the variable is taken in the calculation of IL or CEI, and “detailed” means that the variable is taken in the detailed calculation of IL or CEI.
Table 2. Summary of the results of meta-analysis of the association between exposure determinants and four outcomes with the quality of evidence grouped per health outcome. The overall quality of evidence is based on the GRADE approach, taking into account the risk of bias, inconsistency, impression, and indirectness. The reporting publication bias was not assessed for groups with fewer than 10 studies. (Na: not applicable.).
Table 2. Summary of the results of meta-analysis of the association between exposure determinants and four outcomes with the quality of evidence grouped per health outcome. The overall quality of evidence is based on the GRADE approach, taking into account the risk of bias, inconsistency, impression, and indirectness. The reporting publication bias was not assessed for groups with fewer than 10 studies. (Na: not applicable.).
Exposure Determinant Sorted per Health OutcomeNumber of Studies 1Heterogeneity “I2 Estimate”Summary Estimate of the Association with Health OutcomeEffect95% Confidence IntervalRisk of BiasInconsistencyImprecisionIndirectnessPublication Bias 2Overall Quality of Evidence 3
Cancer60
Active Ingredient1417.171.009Harmful0.992–1.027ModerateNoYesNoYesVery low
Chemical class560.941.053Harmful0.898–1.208HighNoNoNoNaModerate
Type of PPP622.961.033Harmful0.936–1.13ModerateNoYesNoNaLow
Crops944.251.006Harmful0.984–1.028ModerateNoNoNoNaModerate
Task200.856Protective0.715–0.998ModerateNoYesNoNaLow
Frequency200.68Protective0.085–1.275ModerateNoYesNoNaLow
Duration301.096Harmful0.993–1.198ModerateNoYesNoNaLow
Lifetime exposure days2840.670.932Protective0.906–0.959LowNoNoNoYesModerate
Intensity-weighted exposure days3115.570.964Protective0.947–0.982LowNoNoNoYesModerate
Neurotoxicity5
Active Ingredient343.391.014Harmful0.979–1.050HighNoNoYesNaLow
Type of PPP263.441.181Harmful1.109–1.254HighNoNoYesNaLow
Duration201.353Harmful1.160–1.547HighNoNoYesNaLow
Endocrine disruptor—
Diabetes
3
Active Ingredient28.011.034Harmful1.010–1.057HighYesNoNoNaLow
Duration201.192Harmful1.006–1.377HighNoNoNoNaModerate
Frequency213.040.940 0.796–1.084HighNoNoYesNaLow
Endocrine disruptor—
Thyroid
3
Active Ingredient247.231.078Harmful1.047–1.110ModerateYesNoNoNaLow
Intensity-weighted exposure days316.561.090Harmful1.061–1.120ModerateNoNoYesNaModerate
1 Number of studies grouped by heath outcome and determinant of exposure. The quality of evidence was assessed when at least two studies had similar determinant of a health outcome. 2 Funnel plots is available in the Supplementary information: Figures S1–S3. 3 Based on the GRADE, which takes into account the risk of bias, inconsistency, indirectness, imprecision, and publication bias of all studies for a given predictor.
Table 3. Results of the meta-analysis of the association between exposure determinants and cancer when restricting some exposure determinants with another exposure determinants. The significant results are highlighted in italics.
Table 3. Results of the meta-analysis of the association between exposure determinants and cancer when restricting some exposure determinants with another exposure determinants. The significant results are highlighted in italics.
Cumulative Exposure Determinant Sorted per Health OutcomeNumber of StudiesHeterogeneity “I2
Estimate”
Summary Estimate of the Association with Health Outcome95% Confidence Interval
Cancer
Chemical class + Duration201.2850.985; 1.046
Type of PPP + Duration301.0390.996; 1.042
Crops + Duration43.061.0160.959; 1.120
Active ingredient + Duration1111.881.0191.153; 1.418
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

Oltramare, C.; Mediouni, Z.; Shoman, Y.; Hopf, N.B.; Graczyk, H.; Berthet, A. Determinants of Pesticide Exposure in Occupational Studies: A Meta-Analysis. Toxics 2023, 11, 623. https://doi.org/10.3390/toxics11070623

AMA Style

Oltramare C, Mediouni Z, Shoman Y, Hopf NB, Graczyk H, Berthet A. Determinants of Pesticide Exposure in Occupational Studies: A Meta-Analysis. Toxics. 2023; 11(7):623. https://doi.org/10.3390/toxics11070623

Chicago/Turabian Style

Oltramare, Christelle, Zakia Mediouni, Yara Shoman, Nancy B. Hopf, Halshka Graczyk, and Aurélie Berthet. 2023. "Determinants of Pesticide Exposure in Occupational Studies: A Meta-Analysis" Toxics 11, no. 7: 623. https://doi.org/10.3390/toxics11070623

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

Article Metrics

Back to TopTop