Skip to main content

Introduction

Biological monitoring (biomonitoring) in occupational safety and health is the detection of substances (biomarkers) in biological samples of workers, compared to reference values. This article is limited to chemical exposures. Biomonitoring can help in exposure assessment of specific chemicals, characterisation of exposure pathways and potential risks. Biomarkers can detect the exposure, the effect, or reveal susceptibility. Biomonitoring may be interpreted at group or individual level. Most common media are urine and blood. While multitudes of substances can be measured, there are still only limited numbers of validated methods and limit values with scientifically proven background. The first paper on occupational biomonitoring was published in the USA[1], and worldwide mainstreaming started in the 1980s.

Definition of biomonitoring

Human biomonitoring can be defined as the method for assessing human exposure to chemicals or their effects by measuring these chemicals, their metabolites or reaction products in human specimens. Biomonitoring involves measurements of biomarkers in bodily fluids, such as blood, urine, saliva, breast milk, sweat, and other specimens, such as faeces, hair, teeth, and nails [2][3]. In the area of occupational medicine or occupational hygiene, biomonitoring is to be understood as the examination of biological materials of employees for the quantitative determination of hazardous substances, their metabolites or their biochemical and/or biological parameters [4]. Within the occupational context, biomonitoring may help assess actual worker risk, where air monitoring alone may seriously underestimate the total uptake of certain substances. [5]. Characteristics of biomonitoring and workplace air monitoring (also known as environmental monitoring) are summarised in Table 1[6].

Table 1: Comparison of biological and workplace air monitoring

 Biological monitoringWorkplace air monitoring
QuantifyingInternal doseExternal dose
AbsorptionAll routesInhalation only
ConfoundersMetabolic phenotypePersonal protective equipment, substances with similar structure/chemical properties
StandardisationDifficultEasy
InterpretationDifficultModerately difficult
MeasurementIndirect (biomarkers)Usually indirect (dangerous substance)
Ethical issuesImportantNone
VariabilityHighUsually low

Source: Manno, 2010[6]

Classification of biomarkers

A biomarker can be any substance, structure or process that can be monitored in tissues or fluids and that predicts or influences health, or assesses the incidence or biological behaviour of a disease [7]. Biomarkers are early (reversible) signs of exposure, effect or susceptibility with possible adverse health outcome. Biomarkers are classified into three categories depending on their use or the specific context in which the test is being used.

A biomarker of exposure is the substance, or its metabolite, or the product of an interaction that is measured in a compartment or a bodily fluid. Biomarkers of exposure identify and measure chemical residues in tissue or body fluids, metabolites of xenobiotic compounds, or physiological outcomes that occur as a result of exposure [7]. For example lead in blood may fairly represent the recent lead exposure of the individual.

A biomarker of effect is a measurable alteration (biochemical, structural, functional, behavioural, etc.) in an organism that can be associated with an established or potential health impairment or disease. Biomarkers of early disease indicate early biochemical or functional alterations, ranging from natural adaptation to disease. For example the value of Zinc protoporphyrin in blood is increased when lead exposure caused changes in the production of haemoglobin[8].

Biomarkers of genotoxicity (chromosomal aberrations, micronuclei, Comet test) are used to measure exposure to genotoxic chemicals, usually at group level. They are sensitive but not specific indicators and generally inadequate for occupational risk assessment purposes.

A biomarker of susceptibility is the marker of an ability to adversely respond to the challenge of exposure to a chemical. Genes can make certain individuals more vulnerable to toxins such as lead. [9] An example for a susceptibility biomarker is the type of genetic code for 6-aminolevulinic acid dehydratase (ALAD), an enzyme involved in the toxicity of lead that exists in two forms. These effect-modifying factors can be inherent or acquired. Biomarkers of susceptibility are not generally used in routine biomonitoring.

Exposure and biotransformation

The three main exposure pathways to chemicals are inhalation (lungs), dermal (skin) and gastrointestinal (ingestion). Biological monitoring considers the overall systemic exposure (internal dose) and effect (biological effective dose) regardless of the source or pathway. Biomarkers may also reflect circumstances like a change in atmospheric pressure, co-exposures and respiratory rate (e.g. heavy workload), which may all lead to a higher (or smaller) uptake of the substance.

Figure 1: Overview of the process of exposure and biotransformation and possibilities of biomonitoring
Figure 1: Overview of the process of exposure and biotransformation and possibilities of biomonitoring

Once it enters the body the chemical (and its metabolites) can:

  • be distributed among body compartments;
  • undergo various modifications (biotransformation);
  • cause functional changes and diseases;
  • get excreted or deposited.

All these pathways are particular to the chemical substance concerned and may be specific to the individual. They may be influenced by internal or external non-occupational confounders (Figure 1)[10].

Biomarkers can be used effectively if their toxicological background is understood:

  • the fate of the chemical and/or its metabolites in the body (toxicokinetics);
  • the mechanism of the disease/adverse effect (toxicodynamics);
  • the way the individual factor promotes the chemical to cause disease/adverse effect (susceptibility).

In the process of biotransformation the external chemical substances are transformed in the body (metabolism). Enzyme activities are the fundamentals of susceptibility, including those involved in absorption and excretion. Individual varieties in the features of enzymes may result in different speeds and pathways in the metabolism of substances, modifying the dose-response curve. There are many modifiers in biotransformation: sex, age, body mass, co-exposures (workplace, non-occupational: e.g. diet, including fat, alcohol, medication, etc.). Compared to the original substance, its degradation products may be less or more harmful (the latter = activation). These metabolites may be used as biomarkers (of exposure or effect) as well.

Toxicokinetic features explain that many substances (although identifiable in the metabolic process) cannot be used for biomonitoring due to short half-life and/or disappearance from the accessible matrix, e.g. from the blood or the urine. Mathematical (such as physiologically-based toxicokinetic) models simulate metabolic processes in the human body to predict possible biological exposure parameters in a certain exposure scenario. Therefore good knowledge of the the toxicokinetic profile of a substance (absorption, distribution, metabolism (where relevant) and excretion profiles of a substance) are needed for the development of a biomarker and, subsequently, for interpreting the results of biomonitoring [11].

Sampling strategy, chemical analysis

Planning a biomonitoring program for specific purpose starts with the selection of an adequate sampling strategy. Ideally a biomarker of exposure is specific for the exposure concerned (single chemical substance or a group) and reliably detectable, using non-invasive sampling. Timing should be chosen accordingly.

Periodical examinations are set either at fixed intervals or according to the previous biomonitoring results[8].

The analytical method is critical for the validity/reliability of a biomarker: accuracy, precision, reproducibility, recovery, sensitivity and specificity all have high influence to the consistency with the limit and reference values concerned. Several factors may affect the quality of the samples and the measurement of biomarkers: type of the matrix (see below Biologocal matrices), point in time of collection, containers and preservatives and other additives used to stabilise the sample, storage temperature and transport time. Adequate reference material should be applied in adequate concentration range and matrix. Reliable and validated analytical methods must be used supported by internal quality control (IQC) and external quality assurance schemes (EQA)[12].

The success of a biomonitoring program highly depends on good cooperation of partners: workers, the employer, the occupational physician/hygienist, the laboratory.

Biomarker half-lives and timing of sampling

In the different matrices biomarkers have different half-lives, which is the time it takes for the concentration to drop to half. Even a single biomarker may have different half-lives on different time scales: e.g. a quick concentration drop within days, followed by a slow decrease in years. Half-lives of biomarkers and the feasibility of single-compartment or multi-compartment models determine the appropriate sampling strategy: the timing and the frequency of sample collection. For substances with long half-life, during continuous exposure, concentration reaches a certain value that remains stable (equilibrium) and reflects long-term intake. In such a case the timing of sample collection during a working day or even working week is not important. However, enough time must be left to reach equilibrium (e.g. lead, cadmium).

When the half-life is short (a few hours – a day) the concentration varies remarkably during the working day or week, thus timing of sampling is essential. Such a concentration reflects exposure over a short time and may not be representative of average long-term exposure. A meaningful picture of the average exposure may be obtained only from several samples. 

Figure 2: Different half-lives of biomarkers
Figure 2: Different half-lives of biomarkers
Figure 3: Possible pitfalls in biomonitoring of irregular exposures and short-half lives of biomakers
Figure 3: Possible pitfalls in biomonitoring of irregular exposures and short-half lives of biomakers

Examples of different half-lives are illustrated in Figure 2[13]. Usually sampling takes place

  • at the end of shift for short half-live biomarkers (Fig. 2: green);
  • at the end of the work-week for short half-live biomarkers with tendency to cumulate (Fig. 2: orange); and
  • any time for highly cumulating biomarkers with long half-lives (Fig. 2: red).

Samples can also be taken prior to shift or during shift in certain circumstances[13].

As a general rule, sampling is to be undertaken at a time when the inner and the external exposures are in a state of equilibrium which is not to be expected in cases where exposures/activities are short (maintenance, etc.). In such cases, sampling shall be performed at the end of the relevant activity. In case of substances with short half-lives (<4 hours), validity of biomonitoring highly depends on the actual exposure profile during the shift.

Figure 3 illustrates the challenge of biomonitoring for different exposures to substances with short half-lives[13]:

  • If exposure takes place in the first two hours of the shift, subsequent sampling (Fig. 3: green arrow) assesses peak exposure, while end of shift sampling (Fig. 3: red arrow) misses detection.
  • If exposure takes place in the last two hours of the shift, sampling at the end of the shift (Fig. 3: orange arrow) detects peak exposure. If exposure is erroneously reckoned as all-day, the total daily exposure will be overestimated.

 

Biological matrices

Biomonitoring is available in many kinds of biological media (matrices): urine, blood, exhaled air, saliva, sweat, semen, faeces, and several tissues. The appropriate matrix depends on the type of biomarker (exposure, effect, susceptibility) and the type of chemical (parent compound or metabolite, volatile or non-volatile, hydrophilic or hydrophobic, unstable or persistent, etc.). The most important biological samples used in the everyday practice are urine, blood, and occasionally exhaled air.

Urine collection is more readily accepted by workers (easy and not invasive). Concentration in urine usually reflects the mean plasma level of the substance since last urination. Rapidly excreted substances (e.g. solvents) are detected in end-of-shift samples. Timed collected specimens may be more representative. In routine urinary biomonitoring spot samples adjusted for dilution (by creatinine or relative density) are adequate alternatives. Highly diluted or very concentrated urine (creatinine: <0.3, >3.0g/litre; relative density: <1.010, >1.030) are not suitable for such adjustment, thus new specimens should be collected.

Most absorbed substance and active metabolites can be found in blood, which is the second most common biological matrix used in routine biomonitoring. Unlike urine, composition of blood is regulated within narrow limits (e.g. pH, concentrations of natural substances), thus it seldom needs adjustment. Blood sampling is useful for inorganic chemicals (e.g. metals) and for organic chemicals that are poorly metabolised and have a sufficiently long half-life.

Exhaled air analysis is non-invasive and most appropriate to estimate exposure to volatile organic substances (e.g. solvents), although it is much less frequently used.

Other matrices such as Hair, nails, semen, saliva, etc. are matrices for biomonitoring but their use within the occupational context is limited. The table in the WHO Europe report on biomonitoring provides an overview of matrices, as well as their advantages and limitations [2]

Setting of biological limit values

The International Commission on Occupational Health (ICOH) defined Biological Limit Value as the biomarker level that can be directly associated with (the lack of) a biological effect or disease[6]. The European Scientific Committee on Occupational Exposure Limits (SCOEL) defined: ‘A Biological Limit Value (BLV) is a reference value for the evaluation of potential health risk in the practice of occupational health. [...] Exposure concentrations equivalent to the BLV generally do not affect the health of the employee adversely, when they are attained regularly under workplace conditions (8 hrs/day, 5 days/week), except in cases of hypersensitivity.’ ‘It is presented as the concentration in the appropriate biological medium of the relevant agent, its metabolite, or indicator of effect.’[14]. The SCOEL Biological Limit Value (BLV) can be either health-based or exposure-based.

A health based BLV is derived directly from human studies containing data on cohorts with dose response effects or early biological effects. Thus the BLV may not necessarily have a relationship with the Occupational Exposure Limit (OEL) but rather with the levels at which the potential adverse health effects are observed in the study(ies) [5] Although these values are preferred, the number of such biomarkers is limited. Therefore, another option is to derive the BLV from the OEL on the basis of established correlations between air levels and biomarker level. In that case the BLV is obtained from the corresponding Occupational Exposure Limits (OELs) by matching the ‘mean’ level of a biological index with the corresponding OEL (concentration limit in the workplace air). These values are calculated from studies comparing exposures (OEL) and the corresponding biological concentrations observed. For non-traditional working schedules (not 8 hours/day, 5 days/week) BLVs can be derived from toxicokinetic and toxicodynamic bases. When an OEL serves as protection against non-systemic effects (e.g. respiratory irritation), and also for substances with significant non-inhalatory exposure routes, the BLV is set to avoid systemic effects (e.g. intoxication), and is not derived from the OEL. Whenever the toxicological data cannot support a health-based Biological Limit Values, only a Biological Guidance Value (BGV) might be established. This value represents the upper concentration of the substance or a metabolite of the substance in any appropriate biological medium corresponding to a certain percentile (generally 90 or 95 percentile) in a defined reference population. A value exceeding the BGV might help to identify the need for an expert consideration of the working conditions. Unlike BLVs, BGVs are not health-based and therefore do not set a limit between absence or presence of adverse health effects [15]. In 2014, SCOEL published a List of recommended health-based BLVs and BGVs, which includes BLVs or BGVs for 22 substances [15]. No update of this document is available but SCOEL did include BLVs or BGS in recommendations on specific substances such as beryllium [16].

BLV are comparable to Biological Exposure Indices (BEI values) in the US (ACGIH) and Biological Tolerance Values (BAT values) in Germany. BAT (biological tolerance) values are based either on the relationship between external exposure and internal dose, or between the internal dose and the resulting effect of the substance, using the average of internal dose. Differences between BLV and BAT (Biologischer Arbeitsstoff-Toleranzwert or Biological Tolerance Value by the Deutsche Forschungsgemeinschaft – DFG, Germany) disappeared following the harmonisation of definitions. Initially BATs used to be maximum permissible values[17][18].

Biological Exposure Indices (BEI®) of the American Conference of Governmental Industrial Hygienists (ACGIH) are indirect guidance values based mainly on the correlation between the biomarker concentration and the airborne concentration of the original substance[19].

At European level the directive on the protection of the health and safety of workers from the risks related to chemical agents at work (directive 98/24/EC) provides the basis for setting indicative occupational exposure limit values and binding occupational exposure limit values for workplace air. The directive also provides the basis for setting binding biological limit values. The directive defines a biological limit value as the limit of the concentration in the appropriate biological medium of the relevant agent, its metabolite, or an indicator of effect (art. 2, e)). If a binding BLV is established, all Member States have to establish a corresponding national binding biological limit. These national biological limits must be based on the EU value but may not exceed this value. Also, if a binding BLV exists health surveillance is a compulsory requirement for work with the hazardous chemical in question. However, only one binding BLV exists in the EU to date. It is set for the blood-lead level (PbB) [1]. Exposure to carcinogens and mutagens at work is regulated under the directive on the protection of workers from the risks related to exposure to carcinogens or mutagens at work (directive 2004/37/EC). Annex III lists the limit values for occupational exposure but no biological limit values are provided. But, it is stated under annex II of the carcinogens and mutagens directive that health surveillance of the workers exposed to carcinogens and mutagens must include, where appropriate, biological surveillance [2]. Since only one binding biological limit value exists, there are many differences between member states. An overview of biological limit values in the member states can be found in a report from EU-OSHA, 2009 [22].

Limitations of biomonitoring

Only accurate and reproducible analytical results should be used for biomonitoring and health surveillance. The predictive value of an effect biomarker is the extent to which that particular biomarker is capable of correctly separating subjects with a likelihood of impairment or disease from those without it and it may be influenced by different factors.

Biomarker concentration levels decrease when moving on from exposure to disease: this increases measurement uncertainty. Measuring the actual chemical can limit biological variability, but the advantage is that the measurement is not confounded by intermediary biological steps or by identical metabolites of other reactions. However, measuring the metabolite(s) may be better if the parent chemical is unstable or volatile, or the metabolite is directly involved in the mechanism of toxicity (needs activation).

There may be a substantial variation among values obtained from different workers on the same day and also from day to day for a single worker: this may make dose-response relationships harder to assess.

Biological limit values in occupational medicine are set for healthy adults. There is no general rule for setting biological limit values for workers with certain health impairments. The disease may influence the biomarker level directly (e.g. biomarkers of effect), or the disease may alter the uptake, metabolism or excretion of the chemical. For these workers biomonitoring needs to be reconsidered and an individual approach may be necessary.

Certain vulnerable groups may require stricter limit values: e.g. blood lead in fertile women and young workers[8].

Use of biological monitoring

Only highly specific, validated biomonitoring tools should be used as part of routine medical surveillance programs, especially when individual decisions have to be made. Selection of appropriate biomarkers depends on the targeted aims and possibilities. The benefit to workers is highest if the biomarker is close to the target organ and the test has high predictive value.

Biological monitoring plays a key role in occupational risk assessment, complementing workplace air monitoring. It can be used as a part of medical health surveillance[8]. Biomonitoring can be used to perform or validate risk assessment when other approaches are unavailable or inadequate.

Biological monitoring may assess individual exposure to chemicals by different routes, providing additional information for occupational risk assessment from hazard identification to the control of prevention measures. It can be used to identify exposed workers, providing strong indirect evidence for the presence of the chemical in the workplace. Several factors contribute to the disruption of expected correlations between biomonitoring and workplace air monitoring, but biomonitoring, not workplace air monitoring, actually reflects systemic exposure. Biomonitoring may better assess the degree of recent and, also past exposure to chemicals from all routes and clarify dose-response relationships. Use at compounds with a skin notation may benefit the most, because biomonitoring allows monitoring of dermal uptake of the substance.

Biomonitoring can help in the correct interpretation of doubtful clinical tests if the dose-response relationship is known. However, this has been controversially discussed (see Ethical considerations).

There may be varied consequences of biomonitoring results above the reference value. Depending on the exposure, biomarker concerned and Member State policies it may result in:

  • a more frequent biomonitoring;
  • detailed health surveillance of the affected or all exposed workers;
  • revision of the risk assessment;
  • removal of the worker from the exposure/job.

Furthermore, biological monitoring can be used for risk assessment within the framework of REACH (Regulation (EC) No 1907/2006 on the Registration, Evaluation, Authorisation and Restriction of Chemicals (REACH) [23]. For the registration of chemicals a chemical safety report is required that documents the chemical safety assessment undertaken as part of the REACH registration process. This report is the key source for providing information to all users and it is the basis for the other REACH processes (evaluation, authorisation, restriction) [24]. One of the objectives of the risk assessment for preparing the chemical safety report is to determine the Derived No-Effect Level (DNEL). The DNEL is defined as the level below which no adverse effects are expected based on the current knowledge. DNELs may be expressed also as internal exposure biomarkers (DNELbiomarker). In general, when both internal exposure (HBM) and external exposure monitoring data are available, and effects data corresponding to both types of exposure data are accessible, the most appropriate and/or reliable method should be used for the setting of the DNEL [1] [26][27]. But REACH requirements do not include any incentive to understand the relationship between internal exposure biomarkers and effects. Therefore, the development of knowledge on the relationship between effects and the internal doses of substances is not a common practice in the registration dossiers [27]

Interpretation of biomonitoring results

Figure 4: Interpretation of biomonitoring results at the group and at the individual levels.
Figure 4: Interpretation of biomonitoring results at the group and at the individual levels.

Biomarker concentration is compared to appropriate reference values, but the user should be aware of all the uncertainty of the results and how the reference is defined. A biomarker above the reference value does not mean disease or hazard but potentially higher exposure than the reference population. This, however, may not necessarily mean increased health risk, but only if confirmed with repeated measurements and analysis of circumstances (SCOEL). Collective biomarker data (e.g. BEI®) should be used only for the assessment of exposure at group level while individual data (e.g. BAT) are appropriate for drawing conclusions on personal exposure.

Dose-response relationships of effects (Fig. 4: blue, orange and red curves – scientifically established) can be compared to the cumulated frequency distribution of the biomarker (Fig. 4: green area – containing the values of all actual measurements). The probability to observe a certain effect (blue, orange or red) is achieved by projecting 100% of the cumulated frequency distribution (Fig. 4: grey broken line) to intersect the dose-response curves (Fig. 4: orange, blue broken lines): 80% for blue effect, 35% for orange effect, and 0% for red effect. Using an individual result (Fig. 4: crossed circle), the probabilities (Fig. 4: yellow arrow) can be calculated for a single worker.[10]

Interpretation can be easier if variability factors are taken into consideration: uncovering and explaining variability may become a resource more than a limitation and may provide useful information for the interpretation of the results. Special attention should be paid to the thorough explanation of the use of biomarkers of carcinogenicity, emphasising that they are neither biomarkers nor direct predictors of cancer disease.

Ethical considerations

Individual biomonitoring results are medical data, thus should be handled accordingly. Interpretation of the results is the task of a physician trained in the field of occupational health and informed on the entire situation (workplace exposures and worker's health). Data handling, storage and communication are specified in Directive 98/24/EC[20]and in further European and national legislations. Communication (and interpretation) of individual results should be made only towards the worker concerned. Group data can be communicated towards the employer and workers' representatives. The ethical considerations must be regarded during the entire process of a biomonitoring study.

ICOH’s code of ethics states that ‘Biomarkers must be chosen for their validity and relevance for protection of the health of the worker concerned, with due regard to their sensitivity, their specificity and their predictive value’. Principles of this internationally agreed document are:

  • Biomonitoring should not be used as screening tests or for insurance purposes’.
  • Current knowledge in biomonitoring of susceptibility does not justify job opportunity discrimination of affected workers.
  • Priority should be given to non-invasive (urine) and easily collected sampling (spot). In these cases informed consent is usually not required for routine procedures using validated biomarkers.
  • Invasive tests or tests posing a health risk calls for a risk-benefit analysis, and informed consent of the worker [28].

Information and consultation of workers

According to Directive 98/24/EC, workers have a privileged right to be informed and know of the results of any biological monitoring in the framework of health surveillance [20]. The individual worker shall, at his request, have access to the health and exposure records relating to him personally. Where, as a result of health surveillance:

  • a worker is found to have an identifiable disease or adverse health effect which is considered by a doctor or occupational health-care professional to be the result of exposure at work to a hazardous chemical agent, or
  • a binding biological limit value is found to have been exceeded,

the worker shall be informed by the doctor or other suitably qualified person of the result which relates to him personally, including information and advice regarding any health surveillance which he should undergo following the end of the exposure.

Stricter requirements apply, especially as regards record-keeping and information of workers, when carcinogens or mutagens´ exposure occur, laid out in Directive 2004/36/EC (carcinogens and mutagens at work)[22]. For example, all cases of occupational cancers shall be notified to the competent authority. Records shall be kept for at least 40 years following the end of exposure.

Advantages and disadvantages of biological monitoring

‘Without biomonitoring as a tool ... occupational risk assessment of chemicals would be more uncertain and vague than it is’[6].

A unique advantage of biomonitoring as compared to air monitoring is that it takes into consideration:

  • all exposure routes (including inhalation, ingestion and dermal absorption) and circumstances (physical activity, multiple and uneven exposures);
  • the individual response (variability in absorption/metabolism/excretion).

Biomonitoring is also an adequate tool in the assessment of the efficacy of protective equipment, ventilation and other hygiene measures. Biomonitoring may help estimating past exposure and assessing individual susceptibility. Specific and sensitive biomarkers may enable intervention in due time, thus preventing the development of a disease.

The above mentioned advantages also have some drawbacks. Biomarkers:

  • are usually unable to specify the source of the exposure (occupational or non-occupational);
  • may not be sufficiently specific to a particular chemical;
  • are not suitable for identification of workplace contaminations in general;
  • may be interfered by other chemicals in the biological medium (e.g. medications);
  • are not useful at all for the assessment/monitoring of acute and/or local toxic effects (e.g. irritation);
  • and the provision of samples for biomonitoring may be a burden for workers (e.g. blood samples).

The integrated use of both workplace air and biological monitoring is considered still to be the best approach to individual exposure assessment. Although biomonitoring presents advantages for the risk assessment of chemicals, it is not a common practice. A survey among risk assessors (2017), showed that biomonitoring in most cases, is either not used, or used only to a limited extent for carrying out risk assessments. In the domain of OSH biomonitoring is more often used in comparison with biomonitoring for the general population, but the survey also showed that this is often linked to the legal requirement for monitoring exposure to lead [27].

Future perspectives in biological monitoring

Currently there are only 60-90 chemicals (FIOH) (IPASUM) with validated methodologies in biomonitoring in contrast to the thousands of chemicals used in workplaces. This calls for the development of biomonitoring. There is a strong demand to develop non-invasive sampling methods. Besides urine, exhaled air and saliva may become more common. However, even urine analysis of unchanged solvents is not widespread enough in current occupational applications.

The decreasing trend in occupational exposure levels and better analytical methods enable investigation of unchanged volatile compounds in urine, minor metabolites and adducts (reaction products of foreign compounds and body’s own substances) in matrices. Adducts are products of the interaction between a reactive chemical/metabolite and a target molecule of the body. Currently adducts are mostly used in research studies. Haemoglobin adducts may provide insight on the molecular mechanism of toxicity, and reflect long-term exposure. DNA adducts can be considered as biomarkers of exposure, of effect and of susceptibility too. They are envisaged in the future as markers of carcinogen exposure (but not markers of cancer). The ‘omic’ (genomics, transcriptomics, proteomics, etc.) technologies detect early molecular responses and signals at cellular level, like the change in the profile of gene expression in cells caused by the investigated exposure. Although in the research phase, this methodology is a very promising possibility for future biomonitoring[6].

To improve the knowledge on biomonitoring and gather data, in 2017 the EU project HBM4EU (human Biomonitoring for Europe) started. The project a joint effort of 30 countries, the European Environment Agency and the European Commission, co-funded under Horizon 2020. This project will help to improve biomonitoring methods and provide better evidence of the actual exposure of citizens to chemicals.

References

[1] Badham, C. & Taylor, H. B, 'Lead Poisoning: Concerning the Standards Which Should be Used in Diagnosing this Industrial Disease, Together with a New Method for the Determination of Lead in Urine', Studies in Industrial Hygiene, no. 7, Joint Volumes of Papers Presented to the Legislative Council and Legislative Assembly, New South Wales, vol. 1, 1st Session of the 28th Parliament, 1927, p. 52.

[2] Human biomonitoring: facts and figures. Copenhagen: WHO Regional Office for Europe, 2015. Available at: http://www.euro.who.int/__data/assets/pdf_file/0020/276311/Human-biomonitoring-facts-figures-en.pdf

[3] European Human Biomonitoring platform. Retrieved 10 March 2020 from https://www.hbm4eu.eu/the-project/european-hbm-platform/

[4] BAUA, What is Biomonitoring?. Retrieved 10 March 2020 from https://www.baua.de/DE/Themen/Chemikalien-Biostoffe/Gefahrstoffe/Biomonitoring/Was-ist-Biomonitoring/Was-ist-Biomonitoring_node.html

[5] EU Commission, ECHA, Joint Task Force ECHA Committee for Risk Assessment (RAC) and Scientific Committee on Occupational Exposure Limits (SCOEL) on Scientific aspects and methodologies related to the exposure of chemicals at the workplace. Available at: https://echa.europa.eu/documents/10162/13579/rac_joint_scoel_opinion_en.pdf/58265b74-7177-caf7-2937-c7c520768216

[6] Manno, M., Viau, C., in collaboration with Cocker, J., Colosio, C., Lowry, L., Mutti, A., Nordberg, M. & Wang, S., 'Biomonitoring for occupational health risk assessment (BOHRA)', Toxicology Letters, 2010, pp. 3-16.

[7] Ladeira C., Viegas S., Human Biomonitoring – An overview on biomarkers and their application in Occupational and Environmental Health, Biomonitoring, vol. 3, 2016. Available at: https://www.degruyter.com/view/j/bimo.2016.3.issue-1/bimo-2016-0003/bimo-2016-0003.xml?format=INT

[8] EC – European Commission, ''Health surveillance of workers exposed to lead and its ionic compounds'', Practical guidelines of a non-binding nature on the protection of the health and safety of workers from the risks related to chemical agents at work, 2006, pp. 65-73. Available at: http://bookshop.europa.eu/uri?target=EUB:NOTICE:KE6805058:EN:HTML

[9] Kelada, S.N, Shelton, E., Kaufmann, R. B. & Khoury, M. J., 'Delta-aminolevulinic acid dehydratase genotype and lead toxicity: a HuGE review', ''Am. J. Epidemiol.'', 2001, pp. 1-13. Available at: http://www.ncbi.nlm.nih.gov/pubmed/11427399

[10] Manini, P., De Palma, G. & Mutti, A., 'Exposure assessment at the workplace: Implications of biological variability', Toxicology Letters, 2007, pp. 210-218. Letölthető: https://pubmed.ncbi.nlm.nih.gov/17157456/

[11] Bevan R., Brown T., Matthies F., Sams C., Jones K., Hanlon J., La Vedrine M., Human biomonitoring data collection from occupational exposure to pesticides, EFSA, 2017. Available at: https://www.efsa.europa.eu/en/supporting/pub/en-1185

[12] Angerer, J. & Hartwig, A. (eds.), ''The MAK-Collection for Occupational Health and Safety. Part IV: Biomonitoring Methods. Vol. 12'' WILEY-VCH Verlag GmbH&Co. KGaA, Weinheim, 2010.

[13] Fiserova-Bergerova, V. & Vlach, J., 'Timing of sample collection for biological monitoring of occupational exposure', ''The Annals of Occupational Hygiene'', Vol. 41, 1997, pp. 345-353. Retrieved 22 June 2015, from: http://annhyg.oxfordjournals.org/content/41/3/345.full.pdf+html

[14] SCOEL – Scientific Committee on Occupational Exposure Limits, ''Methodology for the Derivation of Occupational Exposure Limits: Key Documentation (version 7)'', 2013, pp. 32-36. Available at: http://ec.europa.eu/social/BlobServlet?docId=4526&langId=en

[15] SCOEL - Scientific Committee on Occupational Exposure Limits, ''List of recommended health-based biological limit values (BLVs) and biological guidance values (BGVs)'', 2014, Available at: http://ec.europa.eu/social/BlobServlet?docId=12629&langId=en

[16] SCOEL/REC/175 beryllium and inorganic beryllium compounds, Recommendation from the Scientific Committee on Occupational Exposure Limits, 2017. Available at: https://op.europa.eu/en/publication-detail/-/publication/732b94b7-0a1b-11e7-8a35-01aa75ed71a1

[17] Drexler, H., Göen, T. & Schaller, K. H., 'Biological tolerance values: change in a paradigm concept from assessment of a single value to use of an average', International Archives of Occupational and Environmental Health 82, 2008, pp. 139-142.

[18] DFG – Deutsche Forschungsgemeinschaft, 'List of MAK and BAT Values', Biological Tolerance Values. WILEY-VCH Verlag GmbH&Co. KGaA, Weinheim, 2010, pp. 201-205.

[19] ACGIH – American Conference of Governmental Industrial Hygienists, ''2010 TLVs® and BEIs®'', Publication #0110, Cincinnati, 2010, p. 95.

[20] Council Directive 98/24/EC of 7 April 1998 on the protection of the health and safety of workers from the risks related to chemical agents at work, Official Journal of the European Communities L 131 of 5 May1998. Available at: http://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri=OJ:L:1998:131:0011:0023:EN:PDF

[21] Directive 2004/37/EC Carcinogens or mutagens at work of 29 April 2004 on the protection of workers from the risks related to exposure to carcinogens or mutagens at work (sixth individual Directive within the meaning of Article 16(1) Directive 89/391/EEC. Available at: https://osha.europa.eu/en/legislation/directive/directive-200437ec-carcinogens-or-mutagens-work

[22] EU-OSHA – European Agency for Safety and Health at Work, ''Exploratory survey of occupational exposure limits for carcinogens, mutagens and reprotoxic substances at EU Member State levels'', Luxembourg, Office for Official Publications of the European Communities, 2009. Available at: https://osha.europa.eu/en/publications/reports/548OELs

[23] Regulation (EC) No 1907/2006 of the European Parliament and of the Council of 18 December 2006 concerning the Registration, Evaluation, Authorisation and Restriction of Chemicals (REACH), establishing a European Chemicals Agency, amending Directive 1999/45/EC and repealing Council Regulation (EEC) No 793/93 and Commission Regulation (EC) No 1488/94 as well as Council Directive 76/769/EEC and Commission Directives 91/155/EEC, 93/67/EEC, 93/105/EC and 2000/21/EC, OJ L 396, 30 December 2006. Available at:http://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri=CELEX:32006R1907:en:NOT

[24] ECHA, Chemical Safety Report. Retrieved at 12 March 2020 from https://echa.europa.eu/regulations/reach/registration/information-requirements/chemical-safety-report

[25] ECHA, Guidance on information requirements and chemical safety assessment, Chapter R.8: Characterisation of dose [concentration]-response for human health, 2012. Available at: https://echa.europa.eu/documents/10162/13632/information_requirements_r8_en.pdf/e153243a-03f0-44c5-8808-88af66223258

[26] Baua, Biomonitoring and REACH. 

[27] Louro H., Heinälä M., Bessems J., Buekers J., Vermeire T., Woutersen M., van Engelen J., Borges T., Rousselle C., Ougier E., Alvito P., Martins C., Assunção R., Silva M., Pronki A., Schaddelee-Scholten B., Del Carmen Gonzalez M., de Alba M., Castaño A., Viegas S., Humar-Juric T., Kononenko L., Lampen A., Vinggaardo A., Schoeters G., Kolossa-Gehring M., Santonen T., Human biomonitoring in health risk assessment in Europe: Current practices and recommendations for the future, International Journal of Hygiene and Environmental Health, vol. 222, issue 5, June 2019, pp. 727-737. Available at: https://www.sciencedirect.com/science/article/pii/S1438463919301543?via%3Dihub

[28] ICOH – International Commission on Occupational Health, ''International Code of Ethics for Occupational Health Professionals'', Third edition, 2014, Available at: http://www.icohweb.org/site/multimedia/code_of_ethics/code-of-ethics-en.pdf

Further reading

EU-OSHA – European Agency for Safety and Health at Work, Dangerous substances e-tool. Available at: https://healthy-workplaces.eu/en/tools-and-publications/ds-e-tool

Aitio, A., Bernard, A., Fowler, B. A. & Nordberg, G. F., Handbook on the toxicology of metals, Academic Press, Burlington, 2011, pp. 65-77. Partly available at: http://books.google.hu/books?id=nKulgztuzL8C&dq

APIS Air Pollution Information System (2014). Retrieved 22 June 2015, from: http://www.apis.ac.uk/overview/issues/overview_biomonitoring.htm

BAuA – Bundesanstalt für Arbeitsschutz und Arbeitsmedizin (2015). Biomonitoring. Retrieved 22 June 2015, from: http://www.baua.de/de/Themen-von-A-Z/Gefahrstoffe/Biomonitoring/Biomonitoring.html

Biomarkers and Risk Assessment: Concepts and Principles (WHO – International Programme for Chemical Safety), ""Environmental Health Criteria""; 155, Report, Geneva, 1993. Retrieved 22 June 2015, from: http://www.inchem.org/documents/ehc/ehc/ehc155.htm

Biomarkers In Risk Assessment: Validity and Validation (United Nations Environment Programme, the International Labour Organization, the World Health Organization). Retrieved 22 June 2015, from: http://www.inchem.org/documents/ehc/ehc/ehc222.htm

Bolt, H. M. & Thier, R., 'Biological monitoring and Biological Limit Values (BLV): The strategy of the European Union', Toxicology Letters 162, 2006, pp. 119-124.

Consortium to Perform Human Biomonitoring on a European Scale (COPHES). Retrieved 22 June 2015, from: http://www.eu-hbm.info/

Eurotox – Federation of European Toxicologists & European Societies of Toxicology (2011). Carcinogenesis Speciality Section. Retrieved 22 June 2015, from: https://www.eurotox.com/carcinogenesis/

HPM4EU- platform. Retrieved 12 March 2020 from https://www.hbm4eu.eu

HSE, Biological monitoring in the workplace. Available at: https://www.hse.gov.uk/pubnS/priced/hsg167.pdf

SCOEL – Scientific Committee on Occupational Exposure Limits (no date). Retrieved 22 June 2015, from: http://ec.europa.eu/social/main.jsp?catId=148&intPageId=684&langId=en

SCOEL – Scientific Committee on Occupational Exposure Limits, Explanation of Key Terms, 2009. Retrieved 22 June 2015, from: http://ec.europa.eu/social/BlobServlet?docId=3880&langId=en

Select theme

Contributor

Karla Van den Broek

Prevent, Belgium

Karla Van den Broek

Prevent, Belgium

Naray Miklos

Klaus Kuhl

Juliet Hassard

Birkbeck, University of London, United Kingdom.
Ellen Schmitz-Felten
Ferenc Kudasz

Frans Jongeneelen