+ All Categories
Home > Documents > DoReMi - Low Dose Research towards Multidisciplinary ... · The aim of CURE was to elaborate a...

DoReMi - Low Dose Research towards Multidisciplinary ... · The aim of CURE was to elaborate a...

Date post: 08-Nov-2018
Category:
Upload: lehanh
View: 216 times
Download: 0 times
Share this document with a friend
77
1 DoReMi - Low Dose Research towards Multidisciplinary Integration Deliverable D5.17: Report for an integrated (biology-dosimetry-epidemiology) research project on occupational Uranium exposure (Task 5.8 CURE Final report) Due date: Month 60 Actual submission date: Month 63 (2 March 2015) Status: Final Nature Report Dissemination level RR = Restricted to a group specified by the consortium (including the Commission Services) Lead beneficiary IRSN Authors Olivier LAURENT (IRSN), Maria GOMOLKA (BfS), Richard HAYLOCK (PHE), Eric BLANCHARDON (IRSN), Augusto GIUSSANI (BfS), Will ATKINSON (Nuvia), Sarah BAATOUT (SCKCEN), Derek BINGHAM (AWE), Elisabeth CARDIS (CREAL), Janet HALL (Institut Curie), Ladislav TOMASEK (SURO), Dominique LAURIER (IRSN) Contributors Sophie ANCELET, Christophe BADIE, Gary BETHEL, Jean-Marc BERTHO, Richard BULL, Cécile CHALLETON de VATHAIRE, Rupert COCKERILL, Estelle DAVESNE, Damien DRUBAY, Teni EBRAHIMIAN, Hilde ENGELS, Nora FENSKE, Michael GILLIES, James GRELLIER, Stephane GRISON, Yann GUEGUEN, Sabine HORNHARDT, Chrystelle IBANEZ, Sylwia KABACIK, Lukas KOTIK, Michaela KREUZER, Anne-Laure LE BACQ, James MARSH, Dietmar NOSSKE, Jackie O'HAGAN, Eileen PERNOT, Matthew PUNCHER, Roel QUINTENS, Estelle RAGE, Tony RIDDELL, Laurence ROY, Eric SAMSON, Maamar SOUIDI, Michelle TURNER, Nina WEILAND, Sergey ZHIVIN Approval WP5 Leader Simon Bouffler: 27 February 2015
Transcript

1

DoReMi - Low Dose Research towards Multidisciplinary Integration

Deliverable

D5.17: Report for an integrated (biology-dosimetry-epidemiology) research project on occupational Uranium exposure

(Task 5.8 CURE Final report)

Due date: Month 60

Actual submission date: Month 63 (2 March 2015)

Status: Final

Nature Report Dissemination level

RR = Restricted to a group specified by the consortium (including the Commission Services)

Lead beneficiary IRSN

Authors

Olivier LAURENT (IRSN), Maria GOMOLKA (BfS), Richard HAYLOCK (PHE), Eric BLANCHARDON (IRSN), Augusto GIUSSANI (BfS), Will ATKINSON (Nuvia), Sarah BAATOUT (SCK•CEN), Derek BINGHAM (AWE), Elisabeth CARDIS (CREAL), Janet HALL (Institut Curie), Ladislav TOMASEK (SURO), Dominique LAURIER (IRSN)

Contributors

Sophie ANCELET, Christophe BADIE, Gary BETHEL, Jean-Marc BERTHO, Richard BULL, Cécile CHALLETON de VATHAIRE, Rupert COCKERILL, Estelle DAVESNE, Damien DRUBAY, Teni EBRAHIMIAN, Hilde ENGELS, Nora FENSKE, Michael GILLIES, James GRELLIER, Stephane GRISON, Yann GUEGUEN, Sabine HORNHARDT, Chrystelle IBANEZ, Sylwia KABACIK, Lukas KOTIK, Michaela KREUZER, Anne-Laure LE BACQ, James MARSH, Dietmar NOSSKE, Jackie O'HAGAN, Eileen PERNOT, Matthew PUNCHER, Roel QUINTENS, Estelle RAGE, Tony RIDDELL, Laurence ROY, Eric SAMSON, Maamar SOUIDI, Michelle TURNER, Nina WEILAND, Sergey ZHIVIN

Approval WP5 Leader Simon Bouffler: 27 February 2015

2

SUMMARY The health effects of chronic exposure to uranium, as it occurs in populations of workers involved in

the nuclear fuel cycle and in the general population, are not well known. Experimental studies on the

effect of exposure to uranium have reported impairments of the cerebral function, genotoxic,

nephrotoxic and other biological effects, but the implications of these findings to human health are

not clear. Beside, a few epidemiological studies reported increases in lung, lymphatic/hematopoietic

cancers and cardiovascular diseases associated with occupational exposure to uranium, but most

available studies are descriptive or suffer from other limitations. New studies borrowing strengths

from modern biology approaches on the one hand, and from large epidemiological datasets on the

other hand, will have better potential to improve the characterization of the biological and health

effects of chronic exposure to low doses of uranium.

The CURE (Concerted Uranium Research in Europe) project was a 18-month concerted action funded

by the European Network of Excellence DoReMi (http://www.doremi-noe.net/). The aim of CURE

was to elaborate a collaborative research project on the biological and health effects of uranium

contamination, integrating epidemiology, biology/toxicology and dosimetry. It notably aimed to

evaluate the feasibility of a molecular epidemiology approach in cohorts of workers exposed to

uranium.

Existing cohorts of workers exposed to uranium (miners and nuclear industry employees involved in

uranium processing) in Belgium, Czech Republic, France, Germany, and the United Kingdom, plus

existing competences in epidemiology, biology and dosimetry among CURE partners, provided a

unique opportunity to develop a coherent multidisciplinary research project within a collaborative

framework. Contacts have been established with other teams involved in similar research projects

within and outside the European Union (e.g.: Russia, Kazakhstan, USA).

Based on the results of this concerted action, a large scale integrated collaborative project will be

proposed to improve the characterization of the biological and health effects associated with

uranium internal contamination in Europe. In the future, it might be envisaged to extend

collaborations with other countries outside the European Union, to apply the proposed approach to

other internal emitters and other exposure situations of internal contamination, and to open the

reflections to other disciplines interested in the effects of internal contaminations by radionuclides.

3

CONTENTS

1. BACKGROUND ................................................................................................................................. 5

1.1. Health effects of internal contaminations by radionuclides ......................................................... 5

1.2. Uranium: a major and widespread radionuclide .......................................................................... 5

1.3. Evidence of uranium effects from experimental studies .............................................................. 6

1.4. Evidence of uranium effects from epidemiological studies .......................................................... 8

1.5. Evidence of uranium effects from molecular epidemiology ......................................................... 9

1.6. Room for improvement ............................................................................................................. 10

1.6.1. Epidemiology .................................................................................................................................... 11

1.6.2. Molecular epidemiology .................................................................................................................... 11

2. OBJECTIVES OF THE CURE PROJECT .............................................................................................. 14

3. PROJECT MANAGEMENT AND TASKS COMPLETED ...................................................................... 14

3.1. General characteristics .............................................................................................................. 14

3.2. Partners ..................................................................................................................................... 14

3.3. Work packages .......................................................................................................................... 17

3.4. Tasks completed ........................................................................................................................ 17

4. PROPOSED INTEGRATED PROTOCOL ............................................................................................ 20

4.1. Epidemiologic cohorts and subsets for analyses ........................................................................ 21

4.1.1. Cohorts for broad base analyses ........................................................................................................ 21

4.1.2. Cohort subsets for dose response analyses ........................................................................................ 23

4.1.3. Cohort subsets proposed for molecular epidemiology ........................................................................ 24

4.2. Biological protocol for a molecular epidemiology study ............................................................. 26

4.2.1. Proposed biomarkers ........................................................................................................................ 27

4.2.2. Biospecimen to be sampled ............................................................................................................... 28

4.2.3. Questionnaire ................................................................................................................................... 32

4.2.4. Information sheet and consent form .................................................................................................. 33

4.2.5. Logistic strategy for the collection of biospecimens in pilot cohorts..................................................... 33

4.3. Dosimetric protocol ................................................................................................................... 41

4.3.1. Introduction ...................................................................................................................................... 41

4.3.2. Biokinetic and dosimetric models ...................................................................................................... 42

4.3.3. Parameters of exposure .................................................................................................................... 43

4.3.4. Monitoring data ................................................................................................................................ 46

4.3.5. Assessment procedure ...................................................................................................................... 48

4.4. Methods for statistical analyses ................................................................................................. 51

4.4.1. Broad base analyses .......................................................................................................................... 51

4.4.2. Dose-response analyses .................................................................................................................... 52

4.4.3. Analysis of biological information ...................................................................................................... 53

4.5. Characterization and propagation of uncertainties .................................................................... 54

4.5.1. Introduction ...................................................................................................................................... 54

4

4.5.2. Characterizing the best estimates and probability distributions of model parameters and other relevant uncertainty sources .......................................................................................................................................... 55

4.5.3. Testing the feasibility to account for the uncertainty inherent in estimates of dose when estimating radiation-induced disease risk .......................................................................................................................... 56

4.5.4. Creating a synthetic cohort as a tool for testing the proposed methodologies ..................................... 57

5. CONCLUSIONS ............................................................................................................................... 58

REFERENCES........................................................................................................................................... 62

LIST OF PARTICIPANTS TO THE CURE PROJECT ...................................................................................... 70

LIST OF INVITED EXPERTS ...................................................................................................................... 71

GLOSSARY .............................................................................................................................................. 72

ACKNOWLEDGEMENTS.......................................................................................................................... 73

ANNEXES ................................................................................................................................................ 74

5

1. BACKGROUND

1.1. Health effects of internal contaminations by radionuclides The assessment of long term health effects resulting from exposure to ionizing radiation is primarily

based on risk models developed from the Life Span study (LSS) of atomic bomb survivors who were

exposed to external photon and neutron radiation in Hiroshima and Nagasaki in 1945 (ICRP, 2007;

NRC, 2006; UNSCEAR, 2008a). By contrast, the major portion of the effective dose delivered to the

general population results from radiation emitted by radionuclides located inside the body after

internal contamination (UNSCEAR, 2008a). Many large populations of radiation workers are also

exposed internally to alpha radiation emitted by radionuclides such as uranium or plutonium.

Currently, estimates of risks associated with internal exposure to alpha radiation are derived from

the risk models based on LSS data, by way of applying a radiation weighting factor (wR) of 20 to the

absorbed alpha dose to account for the experimentally observed difference in risk (per unit absorbed

dose) for alpha particles as compared to photons (ICRP, 2007). However, there are concerns over the

reliability and accuracy of the conversion of risk between these very different types of exposure.

In 2009 the EURATOM-convened High Level and Expert Group (HLEG) considered the limited

knowledge about the effects of internal contamination by radionuclides to be a key scientific issue

for the purpose of radiation protection policy (HLEG, 2009). Subsequently, the effects of internal

contamination have been considered as a cross-cutting issue by European platforms in the field of

radiation research: MELODI (www.melodi-online.eu), EURADOS (http://www.eurados.org), NERIS

(http://www.eu-neris.net) and ALLIANCE (www.er-alliance.org).

The DoReMi network of excellence supported by the European Commission (http://www.doremi-

noe.net/) also identified the gaps in the knowledge of the effects of internal contamination as key

issues for radiation protection research. A workshop organized as part of DoReMi task 5.5 concluded

that further radiobiological research on this topic as well as epidemiological studies in populations

directly exposed to internal emitters are essential (Laurier et al., 2012). This research would allow

the assumptions within the current radiation protection system with respect to risks from internal

emitters (ICRP, 2007) to be evaluated and drive any future improvements to this system. This

workshop also concluded that whenever possible, radiobiological and epidemiological research on

the study of internal contamination should be integrated into the framework of molecular

epidemiology studies nested within established cohorts. Uranium miners and workers were

identified as populations of major interest for the characterization of risks associated with internal

contamination (Laurier et al., 2012), notably to assess the validity of the radiation weighting factor

for alpha emitters (e.g.: (Rage et al., 2012)).

1.2. Uranium: a major and widespread radionuclide Uranium is a naturally occurring radioactive element (symbol U, atomic number 92). Natural uranium

is a mixture of three isotopes: by mass, 238U is the most abundant (99.275%), while 235U and 234U

represent 0.72% and 0.005%, respectively. All three isotopes exhibit the same chemical properties,

but have different radioactive characteristics: 238U has low specific activity, with a half-life of 4.5x109

years, whereas 235U and especially 234U are more radioactive, with half-lives of 7.0x108 and 2.5x105

years, respectively (UNSCEAR, 2008b). In natural uranium approximately 49% of the activity is

produced by 234U and 49% by 238U, with only 2% from 235U. Food and drinking water are the main

6

sources of exposure to natural uranium isotopes among the general population (Ansoborlo et al.,

2015; ATSDR, 2013).

Uranium has important industrial applications since uranium ore is the base material for the

production of nuclear fuel. Uranium is present in various chemical forms and isotopic compositions

at all stages of the nuclear fuel cycle. This cycle includes industrial processes such as mining, milling,

concentration, conversion and enrichment of uranium, fuel fabrication and reprocessing (see annex

1). Most civil reactors use enriched uranium, characterized by a higher proportion of 235U (2 to 5%)

than natural uranium (UNSCEAR, 2008b). Even higher degrees of enrichment can be encountered, for

instance during the production of nuclear weapons (ATSDR, 2013). The industrial process of

enrichment separates natural uranium into enriched uranium and depleted uranium, a by-product

characterized by a lower percentage of 235U (about 0.3%) than natural uranium. Depleted uranium

has been used in various ways, notably as counterweights or ballast in aircraft, shielding and military

applications (tank armor, munitions). Enriched uranium is more radioactive than natural uranium

owing to its higher percentages of 235U and 234U. Conversely, depleted uranium is less radioactive

than natural uranium since it contains a lower proportion of these two isotopes. Other, man-made

(artificial), isotopes of uranium can also be encountered at specific stages of the fuel cycle, such as 232U while handling reprocessed uranium (Guseva Canu et al., 2011).

In summary, interest in the incorporation of uranium into the body is justified by the widespread

exposure of the general population to natural uranium (Ansoborlo et al., 2015; ATSDR, 2013; Canu et

al., 2011) and by the more localized exposure to depleted uranium in settings where conflicts have

occurred (Krunic et al., 2005). In addition, there are many occupational settings in which workers are

exposed to uranium in various forms: natural, depleted, enriched, reprocessed (Laurier et al., 2012).

Since uranium is both a radionuclide and a heavy metal, it has the potential to exert both radiological

and chemical toxicities.

1.3. Evidence of uranium effects from experimental studies Uranium has been found to be genotoxic in both in vitro and in vivo experiments (ATSDR, 2013), even

in its depleted form which generates little alpha radiation (Bal et al., 2011; Stearns et al., 2005). A

recent study showed that depleted and enriched uranium have different genotoxic profiles: depleted

uranium has a low clastogenic but a high aneugenic potential, whereas enriched uranium has a high

clastogenic potential, suggesting additional radiotoxicity on top of chemical toxicity (Darolles et al.,

2010). These genotoxic properties clearly justify further investigations of the association between

uranium exposure and cancer.

In rats, inhalation of depleted uranium has been linked with the formation of DNA strand breaks in

broncho-alveolar lavage cells, possibly as a consequence of uranium-induced inflammation and

oxidative stress (Monleau et al., 2006). Also in rats, chronic inhalation of uranium ore dust was linked

with primary malignant and non-malignant lung tumor formation and although the malignant tumor

risk was not directly proportional to dose, it was directly proportional to dose rate (Mitchel et al.,

1999). In dogs, uranium inhalation was related to neoplasia and fibrosis in the lungs (Leach et al.,

1973). Although only limited experimental data are available on the effects of uranium inhalation on

the lung, their findings suggest the need for further experimental and epidemiological studies. This is

especially important since inhalation is a major route of exposure to uranium in the workplace.

7

Once absorbed, uranium is deposited throughout the body and the highest levels are found in the

bones and kidneys (ATSDR, 2013). Many animal studies have reported damage to the kidney caused

by acute exposure to uranium (Gueguen and Rouas, 2012), which is considered to be the main target

organ for uranium toxic effects under such exposure conditions (ATSDR, 2013). Experimental studies

on the effect of chronic exposure to uranium in kidney have reported mixed findings. Some reported

nephrotoxic effects (Gilman et al., 1998; Zhu et al., 2009), whereas others unexpectedly did not find

such effects (Poisson et al., 2014).

The brain does not accumulate large quantities of uranium. However, some studies in rats have

reported associations between uranium exposure and impairments in cerebral function, suggesting

that the brain is an organ at risk for this nuclide (Houpert et al., 2005; Lestaevel et al., 2005).

The potential effects of uranium on the cardiovascular system have rarely been evaluated in

experimental studies. However, this question is of importance because of the relationships that have

been demonstrated between impaired renal function (which again, may be caused by uranium

(ATSDR, 2013)) and cardiovascular diseases (Currie and Delles, 2013). In addition, the inhalation of

particulate matter of less than 10 µm aerodynamic diameter is an established risk factor for

cardiopulmonary diseases (Araujo and Nel, 2009). Uranium bearing particles of such diameters are

encountered in uranium mines (Marsh et al., 2012) and at all stages of the nuclear fuel cycle

(Ansoborlo et al., 2002).

Further biological effects of uranium exposure have been identified by experimental research,

including oxidative stress, damage to the skin, impaired bone formation or reproductive function

(ATSDR, 2013).

A recent study in rats chronically exposed to low doses of uranium via drinking water showed that a

metabolomics approach using urine samples allowed the correct classification of exposed and non-

exposed animals (Grison et al., 2013). This suggests that metabolomics might be a useful tool to

identify new biomarkers (“fingerprints”) for uranium exposure in humans, as well as to identify

metabolic pathways activated by uranium exposure. This might in turn lead to the identification of

further biological functions and systems perturbed by uranium exposure, which would justify

complementary investigations of potentially related biological and health effects in epidemiological

cohorts.

In summary, it is important to emphasize that experimental studies on natural or depleted uranium

(both being only weakly radioactive, i.e. showing low specific activity) have reported significant

biological effects including genotoxicity, renal toxicity, lung damage and neuro-physiological

perturbations. This suggests that uranium, as a heavy metal, may cause harm by chemical toxicity,

aside from any radiation effects. In addition, experimental research confirmed that radiotoxicity can

occur in addition to chemical toxicity, especially when exposure is to enriched or reprocessed

uranium (Grignard et al., 2008; Houpert et al., 2005).

There are however, considerable uncertainties in the extrapolation of the biological effects at low

doses observed in animals, to possible disease risks in humans (ATSDR, 2013). These uncertainties

are related 1) to possibly different biological responses induced by uranium in animals and in humans

and 2) to the relations of such biological responses with disease risk quantification (e.g., the

predictive value of early biological effects on subsequent disease risk).

8

It is therefore important to evaluate if exposure to uranium is associated with biological and health

effects in humans. The influence of different mixtures of uranium isotopes also appears worth

examining in this frame, in order to disentangle chemical from radiological effects (Zhivin et al.,

2014).

1.4. Evidence of uranium effects from epidemiological studies Cohorts of uranium miners and other workers have been identified as populations of priority interest

for the study of uranium health effects (Laurier et al., 2012). Therefore the present section will focus

on evidence from epidemiological studies conducted in occupational settings. Studies conducted in

non-occupational settings (e.g., in populations living around uranium processing sites) have mostly

employed ecological designs, and are therefore minimally informative. Molecular epidemiology

studies incorporating biomarkers will be reviewed in section 1.5.

Underground uranium miners are exposed to uranium ore dust and are therefore of interest to study

the health effects of uranium exposure. However, previous analyses in these miners predominantly

investigated the health effects related to the exposure to radon (a decay product of uranium or

thorium) (NRC, 1999), whereas the associations between uranium exposure and health outcomes

have been more rarely studied (e.g.: (Kreuzer et al., 2006; Rage et al., 2014)).

The doses from alpha radiation to the lung from uranium and other long-lived radionuclides (LLR)

present in uranium ore are indeed negligible when compared to those from radon exposure (Marsh

et al., 2012). This is particularly true for miners who worked before the implementation of radiation

protection standards (especially ventilation) in mines (Kreuzer et al., 2013; Rage et al., 2014;

Tomasek, 2012). In addition, potentially high correlations between lung doses from radon and from

long-lived radionuclides raise concerns about the possibility of disentangling the effect of uranium

exposure on lung cancer from that of radon exposure.

Non-lung cancer health outcomes are less likely to be affected by any confounding effect of radon

exposure, because doses from radon to the non-respiratory organs where uranium may accumulate

are very low (Marsh et al., 2012). By comparison, doses from LLR to some non-respiratory organs or

tissues (e.g.: kidney, red bone marrow, liver) may be non-negligible (Laurier et al., 2009). In addition,

the chemical toxicity of the uranium dust or other toxic effects resulting from the inhalation of

uranium bearing particles (which might be related to their chemical composition or to other, non-

radiological physical properties such as size (Schlesinger et al., 2006)) might increase the risk of

diseases other than lung cancer (e.g. including, but not necessarily limited to, non-cancer diseases of

the lung and kidney, and circulatory diseases). The associations between uranium exposure and

outcomes other than lung cancer have been evaluated in a few studies of uranium miners (Kreuzer et

al., 2006; Rage et al., 2014), but clearly deserve further investigation.

Epidemiological studies focusing on the health effects of uranium exposure at later stages of the fuel

cycle, where radon exposures are extremely low, have also been performed (ATSDR, 2013; Canu et

al., 2008; Zhivin et al., 2014). These studies included individuals engaged in uranium mills (Boice et

al., 2008; Kreuzer et al., 2014; Pinkerton et al., 2004)) and other uranium processing nuclear

industries such as those located at Port Hope, Canada (Zablotska et al., 2013), Fernald, Ohio, USA

(Ritz, 1999a; Silver et al., 2013), Mallinckrodt, Missouri, USA (Dupree-Ellis et al., 2000), Oak Ridge

National Laboratory, Tennessee, USA (Richardson and Wing, 2006; Yiin et al., 2009), Paducah,

Kentucky, USA (Chan et al., 2010; Figgs, 2013), Rocketdyne, California, USA (Boice et al., 2011),

9

AREVA NC Pierrelatte, France (Guseva Canu et al., 2012; Guseva Canu et al., 2011), Springfields and

Capenhurst, UK (McGeoghegan and Binks, 2000a; McGeoghegan and Binks, 2000b).

However, most of these previous studies were limited by various factors (Canu et al., 2008; Zhivin et

al., 2014):

The statistical power of individual studies conducted in specific settings was limited by

relatively low numbers of workers included (usually a few hundred or thousands) and

sometimes short follow-up times at the time of publication. Because of the often moderate

to low level of exposures (and related low expected excess incidences of relevant diseases),

the numbers of workers and length of follow-up might not have been sufficient to provide

the power needed to detect statistically significant effects.

Reconstructing retrospective exposures for large populations of workers is challenging and as

a consequence many studies used indirect approaches for quantifying exposure. Individual

dose assessment based on urine analysis and other results of individual monitoring for

uranium (faeces, lung or whole body counts) would provide more accurate estimates and

thus increase the ability of studies to detect excess risk (Boice et al., 2006). In addition, most

analytical studies could not take into account the physical and chemical properties of

uranium (isotopic composition, chemical speciation, aerodynamic diameter of particles, and

resulting solubility). These properties may modify the mass and activity of the uranium

reaching different organs, and therefore might determine whether toxic effects of

radiological or chemical nature occur (Guseva Canu et al., 2011).

Finally, only a few studies have collected information on occupational exposures other than

uranium (e.g. (Chan et al., 2010; Guseva Canu et al., 2011; Silver et al., 2013) or on lifestyle

risk factors such as smoking (e.g.(Dupree et al., 1995)). If such factors are correlated with

uranium exposure, they might have biased risk estimates in studies which did not consider

them in the analysis.

As a result, individual epidemiological studies conducted so far do not provide reliable evidence on

the potential health risks associated with uranium exposure (Zhivin et al., 2014). Nevertheless, some

studies are suggestive of biologically plausible positive associations between uranium exposure and

lung cancer (Guseva Canu et al., 2011; Ritz, 1999b), lymphatic and hematopoietic tumours (Guseva

Canu et al., 2011; Yiin et al., 2009), circulatory diseases (Guseva Canu et al., 2012), and intestinal

cancer (Silver et al., 2013). Larger epidemiological studies with adequate individual dosimetry and

control for confounders are clearly required.

1.5. Evidence of uranium effects from molecular epidemiology So far, research on the biological and health effects of uranium has mostly been disconnected

between experimental and epidemiological studies. However, a few pioneering molecular

epidemiology studies have also been conducted.

Some of these studies investigated biological effects of exposure to depleted uranium in human

populations. These populations include Gulf war veterans exposed through friendly-fire incidents via

wound contamination and inhalation (McDiarmid et al., 2011a; McDiarmid et al., 2011b; McDiarmid

et al., 2013). In these veterans, only relatively weak genotoxic, pulmonary or renal adverse effects

have been associated with depleted uranium exposure so far. Other studies were carried out in

workers involved in the clean-up of conflict zones contaminated with depleted uranium munitions in

10

the Balkans (Milacic, 2008; Milacic and Simic, 2009). In these workers, DNA alterations were found at

higher rates immediately after decontamination than before (Milacic, 2008). Damages to

chromosomes and cells were higher in workers exposed to depleted uranium than in controls groups

of workers (Milacic and Simic, 2009).

Other studies were conducted in populations drinking water naturally contaminated with uranium

(Kurttio et al., 2002; Kurttio et al., 2006; Mao et al., 1995; Selden et al., 2009; Zamora et al., 2009). In

these populations, several biomarkers of kidney function indicated some alterations associated with

uranium concentrations in drinking water (ATSDR, 2013). No indication of serious renal injury was

reported since values of biomarkers generally remained in the normal range (Ansoborlo et al., 2015).

However, a cautious interpretation is warranted since the cross sectional design of these studies did

not allow for a detailed characterization of the chronic effects of protracted, cumulated exposure to

uranium (Canu et al., 2011). Similar studies also reported a positive association between uranium

concentrations in water and bone resorption (as indicated by levels of serum type I collagen carboxy-

terminal telopeptide) (Kurttio et al., 2005) or with blood pressure (Kurttio et al., 2006).

Several molecular epidemiology studies were conducted in uranium miners, but generally with a

focus on radon rather than uranium effects (Brandom et al., 1972; Gomolka et al., 2012; Leng et al.,

2013; Li et al., 2014; Zolzer et al., 2012a; Zolzer et al., 2012b).

A few studies were conducted in other uranium workers (Clarkson and Kench, 1952; Dounce et al.,

1949; Eisenbud and Quigley, 1956; Luessenhop et al., 1958; Martin et al., 1991; Prat et al., 2011;

Thun et al., 1985). Most of them focused on renal biomarkers. The earliest studies failed to detect

nephrotoxic effects of uranium exposure, possibly since the biomarkers investigated were not

sensitive enough (ATSDR, 2013). However a study in uranium millers identified reduced renal

proximal tubular reabsorption in this population, as compared to other workers (Thun et al., 1985).

Another study in uranium workers reported a decrease of urinary osteopontin associated with

uranium exposure, suggesting kidney damage (Prat et al., 2011). Another study focused on

cytogenetic markers in blood lymphocytes and identified increases in asymmetrical chromosome

aberrations and sister chromatid exchanges in workers occupationally exposed to uranium (Martin et

al., 1991). An interesting biobank is being set up in uranium and plutonium workers in Russia

(Takhauov et al., 2015) but no result on the association between uranium exposure and biomarkers

is available yet.

So far, published molecular epidemiology studies conducted in uranium workers have focused only

on a limited number of biomarkers (mainly related to kidney function or cytogenetics) and generally

lacked proper organ dosimetry. Most of these studies have been cross sectional and therefore have

not allowed for a detailed assessment of the long-term effects of protracted exposure to uranium.

1.6. Room for improvement

Overall, in spite of experimental results demonstrating chemical and radiological toxicity of uranium

on living organisms, the biological and health effects of chronic uranium exposure in humans,

particularly those due to occupational exposure to uranium, are not well known. However, through

11

conducting improved epidemiological and molecular epidemiology studies it should be possible to

improve the understanding and quantification of these effects.

1.6.1. Epidemiology

Despite the limitations of previous epidemiological studies regarding the quantification of the health

effects of uranium exposure, cohorts of uranium miners and uranium processing workers were

identified as part of DoRemi Task 5.5 as among the most potentially informative source of data for

the characterization of effects of internal contamination (Laurier et al., 2012). The strengths of these

studies include regular collection of dosimetric data and long-term follow-up of worker populations.

In order to allow for such cohorts to provide better insight into the health effects of uranium

exposure, several limitations of previous studies need to be addressed:

First, existing cohorts should be pooled after verification of their compatibility, in order to

increase the statistical power available for analyses and therefore improve the detection and

quantification of potential uranium-related health effects. Similar pooling and joint analyses

of cohorts have successfully been conducted in other populations of radiation workers and

miners (Cardis et al., 2005; Leuraud et al., 2011; NRC, 1999; Tomasek, 2014).

Second, the doses of cohort subjects should be estimated according to state-of-the-art

methods. Methodologies to enable uncertainty in the dose estimates to be incorporated in

dose response analyses should be developed.

Finally, available information on risk factors other than uranium exposure should be carefully

collected in order to adequately control for potential confounding or to take potential

interactions into account in subsequent epidemiological analyses.

It has been acknowledged that setting up multi-country joint epidemiological studies with reliable

dose estimates and using harmonized methods is a highly desirable, but complex, approach. Such

an approach requires preparatory work between researchers (epidemiologists, dosimetrists and

statisticians) in charge of the cohorts to describe and compare available datasets and methods,

and to define harmonized procedures for successfully pooling their data.

1.6.2. Molecular epidemiology

DoReMi task 5.5. has identified that research on the effects of internal contamination would benefit

from the integration of epidemiology and biology within the framework of molecular epidemiology.

Such integration would have a strong potential to improve the understanding and the quantification

of the biological and health effects of radiation exposure, notably resulting from internal

contamination by uranium and other radionuclides (Laurier et al., 2012; Pernot et al., 2012).

The rationale for integrating measurements of biomarkers in a large cohort study on the effects of

internal contamination by uranium is based upon three specific points: 1. to refine the assessment of

exposure to uranium by using biomarkers of exposure (e.g.: (Grison et al., 2013)) 2. to similarly

evaluate possible confounders by the use of confounder specific biomarkers, if available and 3. to

elucidate the different biological response mechanisms induced by uranium exposure in a human

population by using biomarkers of early and late biological effects (Pernot et al., 2012).

12

If any association is observed between uranium exposure and the risk to develop a disease, point 3)

may provide insight into the physio-pathological processes involved in disease onset. This will help

assess the biological plausibility of the associations observed in epidemiological studies and inform

related judgment about causality. This might also help to identify pre-clinical biomarkers useful for

the surveillance of people exposed to uranium.

Again, it should be possible to overcome the main limitations of previous molecular epidemiology

studies, and to build on recent, as well as future, progresses of techniques for biological analyses:

Although most previous molecular epidemiology studies have been cross sectional, regular

medical check-ups conducted by the occupational medicine service during the workers’

careers have been identified as a unique opportunity for the prospective and repeated

collection of biological samples among workers (Laurier et al., 2012). A biobank centralizing

such samples over many years from a same cohort of individuals would be a valuable

resource for a detailed assessment of the long-term effects of protracted exposure to

uranium, as it is the case for other exposures (Palmer, 2007; Zins et al., 2010). Such a

prospective scheme is also of great interest to identify, or to validate, biomarkers of recent

exposures (e.g.: (Grison et al., 2013)) and their persistence or modifications over time.

In addition, the immediate collection of biological material from workers exposed to uranium

or other sources of radiation years to decades ago is of great interest in order to directly

investigate any persistent biomarkers of past exposures (Gomolka et al., 2012). If specific

diseases occurred in some of these workers, pathological archives (e.g. formalin fixed

paraffin embedded tissue from target organs (Wiethege et al., 1999)) may also be of great

interest to decipher uranium specific fingerprints in diseased tissues, for instance in tumor

tissues from cancer cases.

The need to use the best possible dosimetric estimates was mentioned above for

epidemiology, and this applies to molecular epidemiology as well. An efficient solution to

address this issue is to nest molecular epidemiology studies within established cohorts for

which dosimetric data have already been carefully collected over many years to decades

(Gomolka et al., 2012), or will be. This consideration also applies to many other factors for

which data are already available within cohorts (e.g., potential confounders).

Novel and already established biomarkers, notably originating from animal experiments, may

prove useful to better understand the biological effects of uranium exposure on its different

target organs, tissues and systems in humans. Some of these biomarkers have never been

tested in humans exposed to uranium, but appear worth investigating for instance in

uranium workers, as they might prove to be of interest to occupational health surveillance

before the onset of any disease (e.g., biomarkers of brain damage, inflammation or kidney

dysfunction).

Non-targeted (OMIC) approaches are also promising for the purpose of identifying not only

new biomarkers of exposure, but also biological (e.g., metabolic) pathways potentially

affected by uranium exposure (Grison et al., 2013), and help direct research towards related

pathways, functions/processes, organs and tissues.

Overall, modern biology approaches are developing fast and should lead to the identification

of new biomarkers of high interest in the forthcoming years or decades. Modern biobanking

methods allow for the long term conservation of biological material in suitable conditions for

later analyses of emerging biomarkers (Palmer, 2007; Zins et al., 2010). Diseases potentially

13

related to current uranium exposure might only be diagnosed many years, even decades,

following exposure. Being able to investigate biological samples collected from the time of

exposure to possible disease onset using the latest available techniques for biological

analyses would be an invaluable asset for future research into potential mechanisms of

disease onset.

As for the preparation of pooled epidemiological studies, cooperation and coordination between

researchers (biologists, epidemiologists, dosimetrists and statisticians) is essential, in order to

undertake any successful and informative pilot molecular epidemiology study in a population of

uranium miners or workers. These discussions should result in a good understanding of the

expectations from the study field by each partner and related justifications, as well as the mutual

understanding of each partner’s (and discipline’s) role within the frame of molecular epidemiology

studies. To set up a molecular epidemiology study, the identification of biomarkers useful to study

the effects of uranium exposure in humans is necessary as well as the identification of populations

suitable for the collection of appropriate biospecimens. Valid biomarker typing can only been done in

biological samples of good quality. Therefore the definition of proper study instruments (standard

operating procedures, questionnaire, information and consent sheets), and the setting-up of an

optimal sampling scheme are essential requirements. These have to be a priori verified and tested in

a smaller feasibility study. The agreement and support of all stakeholders (workers, worker

representatives, employers, national ethics committees,) must be obtained before undertaking the

study. The experiences and feedback of other European initiatives in the field must be used

especially from the European Biobanking and Biomolecular Resources Research Infrastructure

(BBMRI) project (bbmri-eric.eu).

14

2. OBJECTIVES OF THE CURE PROJECT

The objective of the concerted action CURE (Concerted Uranium Research in Europe) was to

elaborate a multidisciplinary and collaborative European research project, integrating epidemiology,

biology/toxicology and dosimetry to improve understanding and quantification of long-term

biological and health effects (including both risks of cancer and non-cancer diseases) associated with

uranium contamination.

This general objective included two specific aims:

• To prepare a common protocol for pooled epidemiological analyses of uranium miners and

workers in Europe, that would overcome the limitations of previous studies in the field, in

order to directly estimate the potential health risks associated with uranium exposure.

• To verify the feasibility of a molecular epidemiology approach (building biobanks and

measuring biomarkers) for integration into the risk assessment process and if feasible, to

elaborate a common protocol for the development of a molecular epidemiology study.

A parallel objective of the project was to identify and establish contact with similar research

programs, within and outside Europe.

3. PROJECT MANAGEMENT AND TASKS COMPLETED

3.1. General characteristics

CURE was a concerted action supported by the DoReMi EU FP7 network of excellence

(http://www.doremi-noe.net/) and coordinated by Institut de Radioprotection et de Sûreté Nucléaire

(IRSN, France). It was integrated as task 5.8 of DoReMi. The project duration was 18 months, from

July 1, 2013 to December 31, 2014.

3.2. Partners

CURE gathered together the main organizations in charge of conducting and/or analyzing cohort

studies of miners and other workers occupationally exposed to uranium in Europe, as well as

organizations with recognized expertise in internal dosimetry and/or in radiobiology.

In total 9 partners from 6 Countries (France, UK, Germany, Belgium, Czech Republic, Spain)

participated in the CURE project: Institut de Radioprotection et de Sûreté Nucléaire (IRSN, France),

Bundesamt für Strahlenschutz (BfS, Germany), Public Health England (PHE, United Kingdom), Nuvia

limited (United Kingdom), Atomic Weapons Establishment (AWE, United Kingdom), StudieCentrum

voor Kernenergie • Centre d'étude de l'Energie Nucléaire (SCK•CEN, Belgium) , Státní ústav radiační

ochrany (SURO, Czech Republic), Centre de Recerca en Epidemiologia Ambiental (CREAL, Spain),

Institut Curie (IC, France).

All partners were involved in the reviews conducted in DoReMi WP4 on pertinent cohorts for

radiation protection, in WP5 task 5.5 on cancer risk and internal contamination (Laurier et al., 2012),

15

in WP6 on useful biomarkers for epidemiology (Pernot et al., 2012) and in WP7 Task7.2 on pertinent

studies for low dose cardiovascular risk.

The respective experiences and fields of expertise of the partners involved in CURE and directly

relevant to this project are summarized below:

IRSN has experience in European collaborative projects (e.g.: DoReMi WP7 and WP5-Task5.5,

coordination of the FP6 Alpha-Risk project), in the conduction of epidemiological cohorts studies of

uranium miners and of uranium workers, in internal dosimetry of uranium miners and of uranium

workers, in biodosimetry, in the investigation of biological and toxicological effects of uranium

exposure (Envirhom program) and in advanced biostatistics. Two PhD students at IRSN (Drubay D,

Zhivin S) participated to the CURE project.

BfS has experience in European collaborative projects (Alpha-Risk, DoReMi, Multibiodose, RENEB,

etc), in epidemiological cohorts of uranium miners, in biobanking of radiation exposed cohorts and

radiation sensitive individuals (German Uranium Miners Biobank), in the analyses of biological effects

(Chromosomal analyses [mFISH, Giemsa], gammaH2AX, Comet assay, Proteomics) of radiation

exposure, of radiation sensitivity, biological effectiveness of radiation, and in dosimetry of internal

emitters.

PHE has experience of European collaborative projects (Alpha-Risk, SOUL, EpiRadBio, SOLO). As part

of the SOLO project, PHE performed coordination, dosimetric reconstruction, biological and

epidemiological risk analysis tasks. PHE has expertise in epidemiological cohorts of nuclear workers

(PHE manages both the UK National Registry for Radiation Workers comprising of over 200 thousand

workers and a cohort of over 60 thousand former employees of British Nuclear Fuels Limited), in

internal dosimetry for uranium and plutonium exposed workers and for uranium miners. PHE has

expertise in radiation biology (RISK-IR), for instance in the field of cancer genetics and cytogenetics,

and carries out research into the fundamental mechanisms by which radiation causes cancer.

Nuvia and AWE have experience in European collaborative projects (Alpha-Risk), in epidemiological

cohorts of uranium workers, in the monitoring of workers exposures and in the assessment of

internal doses. Nuvia and AWE are collaborators in the AIRDoseUK project within DoReMi (task

5.5.2.), which will improve the organ doses calculation among United Kingdom Atomic Energy

Authority (UKAEA) workers.

SCK•CEN has experience in European collaborative projects (NOTE, EPI-CT, Alpha-Risk, CEREBRAD,

PROCARDIO, GENRISK-T), in the analysis of biological/molecular effects of radiation exposure,

radiation sensitivity as well as on cancer susceptibility markers and non-cancer diseases using up-to-

date infrastructures. SCK•CEN has the long-term commitment to develop research in low dose

radiation issues, and in epidemiological cohorts of nuclear workers.

SURO has experience in European collaborative projects (Alpha-Risk). SURO has expertise in

epidemiological cohorts of uranium miners, internal dosimetry, radiochemistry, statistics and

occupational medicine. Within DoReMi, SURO is leading the IntEmitUM project (task 5.5.1), which

aims to the integration of dosimetry, biology and epidemiology to estimate cancer risks related to

internal exposure among miners using measurement of uranium in urine.

16

CREAL has experience in European collaborative projects (e.g.: Alpha-Risk, Int-Thyr), in the

epidemiology of nuclear workers and in risk modelling, in molecular epidemiology and biomarkers.

CREAL has experience in the consideration of dose uncertainties and their propagation into risk

estimates (nuclear workers, Chernobyl liquidators). In DoReMi, CREAL is coordinating the cross-

cutting molecular epidemiology group and the reflexions on molecular epidemiology in WP4 and

WP6.

IC has considerable experience in molecular epidemiology studies and biomarkers. In the framework

of DoReMI, IC participates in the cross-cutting molecular epidemiology group.

17

3.3. Work packages

The CURE project included four work packages (WPs):

WP1 (leader: Richard Haylock, PHE) dedicated to epidemiology

WP2 (leader: Eric Blanchardon, IRSN) dedicated to dosimetry

WP3 (leader: Maria Gomolka, BfS) dedicated to biology

WP4 (leader: Dominique Laurier, IRSN) dedicated to the management and general

coordination of the project

In addition, it was decided in March 2014 to create an uncertainty work group (UWG, leader:

Augusto Giussani, BfS), including members from all the main Work Packages of CURE (epidemiology,

dosimetry and biology). The UWG was dedicated to the coordination and harmonization of the

methods for addressing the cross-cutting issue of uncertainties at different steps of the project.

3.4. Tasks completed

The main tasks of each work package and interactions between them are briefly summarized in

Figure 1.

Figure 1. Work packages of the CURE project, main tasks and interactions between work packages

(WPs) and the Uncertainty Work Group (UWG).

A more detailed list of the tasks performed as part of the CURE project is provided below. Because of

the strong interactions between the different WPs and disciplines, many tasks were conducted

18

jointly by two or more WPs. Therefore, many tasks completed could not strictly be grouped by WP,

but the contributions of each WP to the listed tasks are mentioned:

Review of regulatory issues in each country (agreements from ethics committees and

workers’ representatives), both for epidemiology and molecular epidemiology studies. See

(Haylock et al., 2014a) for details (WP1+WP3).

Detailed description of the cohorts of miners and workers to verify their compatibility

(nature of the variables collected, availability, quality and extent of follow-up, quality and

completeness of dosimetric information) and identify the specific strengths of some sub-

cohorts (e.g.: data on potential confounders available either from Job exposure matrices

(JEMs) or from medical files, cancer incidence data). See (Haylock et al., 2014b) for details

(WP1+WP2)

Definition of cohorts or cohort subsets suitable for different kind of epidemiological analyses,

see section 4.1. (WP1 + WP2)

Definition of a common general strategy for molecular epidemiology (i.e.: to perform

biological sampling, biomarker measurements and analyses in a cohort of uranium exposed

workers and proposition of cohort subsets for pilot molecular epidemiology studies, see

section 4.1.3. (WP1 + WP3)

Identification of relevant biomarkers of exposure or of potential early or late effects, of

interest to study the biological and health effects of uranium (Gomolka et al., 2014). Both

targeted biomarkers (selected notably on the basis of previous experimental studies) and

non-targeted biomarkers (OMICs) were considered (WP3).

Identification of biological specimens to be collected for the measurement of the selected

biomarkers, see section 4.2.2. (WP3)

Definition of standard operation procedures (SOPs) for biological samples and biomarkers

(collection, processing, storage, analysis) (Gomolka et al., 2014) (WP3)

Development of instruments for a pilot molecular epidemiology study (Gomolka et al., 2014):

questionnaire (see section 4.2.3.), information sheet and consent form (see section 4.2.4.)

(WP1+ WP3)

Verification of the feasibility of pilot studies in the proposed cohort subsets for molecular

epidemiology: onsite visits, collection of information on field conditions, and comparison

with SOPs, see section 4.2.5. (WP1+ WP3)

Development and adaptation of the logistic strategy in the pilot cohorts to inform the

medical doctors, workers, to collect and transport the biospecimens and to isolate and store

the biological material of interest, see section 4.2.5. (WP1+ WP3)

Identification of centers able to provide or assist in biospecimen sampling, storage and/or specific biomarker testing (Gomolka et al., 2014)(WP3)

Evaluation of the costs of biospecimen sampling, storage and biomarker testing (Gomolka et

al., 2014) (WP3)

Definition of the relevant biokinetic and dosimetric target organs, relevant to the diseases

(see section 4.4.) (WP2+ WP1) and the biomarkers (see section 4.2.1.) of interest (WP2+

WP3)

Detailed evaluation of the availability and quality of monitoring data available in each cohort

for dosimetric calculations (Blanchardon et al., 2014b) (WP1+ WP2)

Analysis of similarities and differences in exposure reconstruction approaches for miners and

workers (Blanchardon et al., 2014a) (WP2)

19

Definition of a dosimetry protocol, harmonizing the dose assessment procedures for similarly

exposed subjects across different countries and sites (Blanchardon et al., 2014a) to allow for

joint epidemiological and molecular epidemiology analyses, see section 4.3. (WP2)

Creation of the uncertainty work group, to address the cross-cutting issue of uncertainties

(UWG = WP1+ WP2 + WP3)

Identification of the potential sources of uncertainties at several steps of the project via the

collective building of a matrix listing identified sources of uncertainties at each step (Giussani

et al., 2014): data collection, dose calculation process, epidemiological and biomarker

analyses (WP1+ WP2 + WP3 within UWG)

Case studies to assess the relative impact of certain sources of uncertainties on dose

calculations (WP2 within UWG)

Prioritization of the sources of uncertainties to be considered (e.g.: to be further

characterized or reduced, whenever feasible) (WP1+ WP2 + WP3 within UWG)

Propositions of methodologies for the propagation of uncertainties in epidemiological

analyses (WP1 within UWG)

Establishment of contacts with other researchers involved in studies of uranium workers or

miners inside or outside Europe, in order to identify their potential synergies with CURE for

future projects (WP4 + WP1 + WP3)

Establishment of contacts with researchers outside the radiation field for input of already

existing knowledge in the biobanking field and identifications of potential synergies with

other projects (WP3+WP4)

Setting up of a CURE web space hosted by DoReMi website (WP4)

General project coordination (WP4)

Early dissemination work : presentations at several workshops and conference (WP4), see

annex 2

20

4. PROPOSED INTEGRATED PROTOCOL

The current protocol was designed with two specific aims:

to set up a joint epidemiological study in order to directly estimate risks from occupational

exposure to uranium, by pooling data from established nuclear worker and miner/miller

cohorts from five countries (United Kingdom, France, Belgium, Czech Republic, Germany).

This would generate datasets with the highest possible statistical power to provide evidence

for the appropriateness of radiation protection standards for people occupationally exposed

to uranium. More generally, this would contribute to improve the knowledge of the health

effects (both cancer and non-cancer) of internal contamination and help assess the

appropriateness of current radiation protection standard regarding internal exposure.

to investigate the feasibility of setting up molecular epidemiology studies. Prospective

molecular epidemiology studies would allow for an improved understanding of the

mechanisms through which uranium might generate health effects and possibly to a better

quantification of related health risks. It might help to identify biomarkers useful to the health

surveillance of workers exposed to uranium. Thus it is important to test the feasibility to

integrate pertinent biomarkers into an epidemiological study.

For each aim, importance has been given to the production and use of the best possible dosimetric

estimates, to the identification of sources of uncertainties at each step of the research protocol, and

to the identification of possible methods for propagating such uncertainties into the analyses.

As a result, the outline of the protocol is divided into the following sections:

Epidemiologic cohorts and subsets for analyses

Biological protocol for a molecular epidemiology study

Dosimetric protocol

Methods for statistical analyses

Characterization and propagation of uncertainties

21

4.1. Epidemiologic cohorts and subsets for analyses

This section presents populations identified as suitable for three different levels of analyses as part of

the CURE project.

A large-scale combined analysis of risks among workers and miners exposed to uranium in

Europe, providing substantial statistical power (“broad base analysis”, e.g. comparison of the

mortality and/or morbidity observed in workers to those observed in the general population).

The aim of this analysis is to identify whether any specific mortality or morbidity profile can be

identified in populations of workers and/or miners exposed to uranium.

Analysis of the association between uranium exposure and health risks (“dose-response

analyses”) among subgroups of workers for which doses are readily available or can be estimated

by mid-2016. Preconditions for pooling are the homogeneity of exposure patterns and the

availability of high quality data to estimate organ doses (individual bioassay data, JEMs). In some

portions of these subgroups selected for dose-response analyses, detailed information on

potential confounders such as smoking, body mass index or occupational exposures other than

uranium are available. This will allow for an assessment of the impact of these factors on dose-

response relationships, as part of sensitivity analyses.

Groups within which the collection of biological samples appears possible and justified, to verify

the feasibility of setting up biobanks of uranium exposed workers. An aim of the CURE project

was to identify, as a result of an agreement between WP1 and WP3, “pilot cohorts for biological

data”, in which feasibility studies have appeared worth conducting.

These populations will be presented in more detail below.

4.1.1. Cohorts for broad base analyses

Six groups of miners and other workers exposed to uranium in Europe have been proposed for

inclusion in a pooled epidemiological study. The detailed presentation of each of these cohorts is

available in CURE deliverable D1.2. (Haylock et al., 2014b). In the current protocol, the presentation

of the proposed joint cohorts will be summarised.

Cohorts of miners and millers will be presented separately from cohorts of other uranium workers

because of the markedly different exposure conditions and the significantly different dose

assessment procedures for these groups (as will be detailed in section “4.3. Dosimetry protocol”

below). For simplicity, in the rest of the report, the term “worker” will exclude miners and millers.

4.1.1.1. Cohorts of miners and millers

Cohorts of uranium miners have been set up in three European countries, namely in Germany (N=

58,982 miners) (Kreuzer et al., 2010), France (N=5,086) (Rage et al., 2014) and in the Czech Republic

(N=7,513) (Tomasek, 2012). These populations (total N>70,000) have been followed-up for several

decades. Researchers involved in these cohort studies will be able to build on previous experiences

of collaboration, for instance as part of the FP6 project Alpha risk, during which subsets of data from

these three cohorts were successfully pooled (Leuraud et al., 2011).

22

As mentioned in the introduction, uranium miners were not only exposed to uranium ore dust but

also to relatively high levels of radon, especially in the early years of underground mining (1940s and

1950s) due to bad ventilation conditions. At that time, absorbed lung doses from inhalation of radon

were appreciably higher compared to lung doses from inhalation of uranium dust. For this reason, it

was decided as part of the CURE project to identify subgroups of cohort members exposed to low

radon concentrations, but still relatively high uranium concentrations, so that radon exposure is

unlikely to obscure potential effects of uranium exposure on lung cancer. These selected populations

include:

Miners who were employed in 1955 or later, thus excluding those who had worked in the

very early years, when the levels of radon concentrations had been extremely high (1946 to

about 1955 or 1958). A large joint cohort of about 40,000 uranium miners or millers

restricted to low radon exposures will be generated by pooling data from Germany (~

29,400), France (~3,400) and the Czech Republic (~7,500). Details are provided in CURE D2.1.

(Haylock et al., 2014b). Such a joint cohort will include miners exposed to uranium ore dust

and German millers additionally exposed to yellowcake (a type of uranium concentrate

powder obtained from leach solutions).

All millers and open pit miners, who never worked underground and had very low radon

exposures (no period restriction was applied). A specific cohort of uranium millers has

already been set up in Germany (Kreuzer et al., 2014) (N=4,054) and efforts are underway to

set up similar cohorts in France (N ~ 1,150) and in the Czech Republic (n=980). This will allow

for a future pooled analysis of mortality risk in millers predominantly exposed to uranium. In

the short term, it will be possible to conduct sensitivity analyses among a group of 6,227

millers and open pit miners who never worked underground. This will include 5,460 German

millers and open pit miners and 767 French open pit miners.

4.1.1.2. Cohorts of nuclear workers

At later stages of the nuclear fuel cycle, other workers have been exposed to uranium as part of

various activities: purification, conversion, enrichment, fuel manufacturing and reprocessing,

decommissioning, recovery and decontamination of effluents and waste, weapon production and

research (see annex 1).

In Europe, these workers have been included in several cohorts. These include three cohorts in the

United Kingdom : British Nuclear Fuels Limited (BNFL) cohort (Gillies and Haylock, 2014), United

Kingdom Atomic Energy Authority (UKAEA) (Atkinson et al., 2007) and Atomic Weapons

Establishment (AWE) (Carpenter et al., 1994; Johnson et al., 1999) cohorts (these later two are both

contained within the SHIELD database). A cohort of workers involved in the nuclear fuel cycle has

also been set up in France: the TRACY cohort (Samson et al., in preparation; Samson et al., 2014). A

cohort of nuclear workers has already been set up in Belgium (Engels et al., 2005), but the overlap

with the population of uranium exposed workers in this country (who were employed by Franco

Belge de Fabrication du Combustible (FBFC)) is only partial as of now.

For the first time, a large joint cohort of about 40,000 nuclear uranium workers will be generated by

pooling data from the UK (~24,500), France (~12,700) and Belgium (~1,650). Such a joint cohort will

23

cover the major stages of the uranium cycle. It will also cover exposures to a large variety of uranium

compounds: U3O8, UO2(NO3)2, (NH4)2U2O7, UO3, UO2, UF4. UF6, UO2F2.

The Belgian cohort of 1,650 workers employed at Franco Belge de Fabrication du Combustible in

Belgium still needs to be set-up using available computerized information.

4.1.2. Cohort subsets for dose response analyses

A dose response analysis will be most effective and informative if it is restricted to high quality data

and if potential for confounding bias is avoided. To this end a subset of data will be selected from

each cohort. Criteria for data inclusion differ between miners/millers and workers cohorts.

4.1.2.1. Cohorts of miners and millers

In these cohorts individual monitoring has usually not been conducted. Exposure in uranium miners

has been predominantly regulated by air sampling, as will be detailed in section 4.3. For all miners,

individual information on exposure to uranium, radon and external gamma radiation is already

available, mostly from ambient measurements and based on detailed job-exposure matrices. Much

individual level information has already been computerized, including internal doses to several

organs (lung, kidney, liver, red bone marrow) which were calculated within the framework of the FP6

Alpha risk project (Marsh et al., 2012). Doses to the stomach are also available for German miners.

The population used for dose-response analyses will be the same as that used for the broad analyses.

Specific sensitivity analyses will be based on the group of 6,227 millers and open pit miners, because

of their high uranium exposure and very low radon exposures. However, for millers the organ doses

have still to be calculated.

4.1.2.2. Cohorts of nuclear workers

In worker populations involved in other stages of the nuclear cycle, the evaluation of occupational

internal exposure to radionuclides has usually been based on individual monitoring of intakes of

radionuclides, as will be detailed in section 4.3. Routine bioassay measurement of radionuclides in

urine, faeces, whole-body or lungs has been conducted on a regular basis, and additional follow-up

measurements have been performed in case of incidents.

For the nuclear worker cohorts, the availability and quality of such data, and the ability to collect or

compute good quality estimates of dose from uranium and other alpha emitting nuclides is therefore

the most important criterion for inclusion in analyses of dose-response relationships. It was

proposed, as part of CURE, to focus on subgroups for which both external and internal dose

estimates could be available by mid-2016.

It is estimated that such data can be obtained for 4,500 workers from France, 24,700 from the UK

(15,000 from BNFL, 3700 from AWE and 6000 from UKAEA – doses in AWE and UKAEA cohorts are

being computed as part of the DoReMi AirDose UK project – task 5.5.2) and 800 from Belgium. In

total, a cohort of 30,000 workers would be available for the purpose of studying dose-response

relationships.

In some portions of this subset (described below), detailed information on potential confounders such as smoking, body mass index, blood pressure or occupational exposures other than uranium are

24

available. This will allow for an assessment the impact such factors on dose-response relationships, as part of sensitivity analyses:

In the TRACY cohort, an effort is underway to computerize individual information on risk

factors available from occupational medical files such as smoking habits, blood pressure,

body mass index, blood sugar and cholesterol, as well as chest X-ray examinations (Garsi et

al., 2014). Repeated information is available for these factors, generally on a yearly or more

frequent basis. Such data will be fully computerized for about 4,500 workers (for which dose

estimates will also be available) by the end of 2016.

Job exposure matrices (JEMs) based on a semi-quantitative approach have already been

developed for several facilities included in TRACY (e.g.: AREVA NC Pierrelatte (Guseva Canu

et al., 2009) and EURODIF (Guseva Canu et al., 2013)). These JEMs cover more than 5,000

workers formerly employed at these plants. They document the exposure of workers to

physical risk factors (e.g. noise, heat, asbestos) and chemicals (e.g.: trichloroethylene,

fluorinated products, aromatic solvents). In addition, these JEMs document the type of

uranium to which workers have been exposed (i.e. details on the degree of enrichment and

chemical form determining solubility) and are therefore of interest to refine dosimetric

estimates and conduct sensitivity analyses on the use of such information for dose

calculation. By mid-2016, a new JEM will be created for the Malvesi plant. In total, about

6500 workers from the TRACY cohort will be covered by JEMs at this time.

A first attempt at performing a JEM in Plutonium workers at the Sellafield plant in the UK is

currently being carried out. The aim in this setting is to improve the information on the

potential exposures of unmonitored workers. If successful it will provide a basis for

evaluating the feasibility to perform similar work on uranium exposed workers to identity the

types of uranium compounds workers were potentially exposed to.

In addition, cancer incidence data are available in the UK cohorts from the beginning of 1971 and will

also be available for the Belgian cohort from the beginning of 1999. This will allow for further

sensitivity analyses of the impact of using mortality vs incidence data when studying the association

between radiation dose and cancer.

4.1.3. Cohort subsets proposed for pilot molecular epidemiology studies

Regarding molecular epidemiology, this section will present groups of individuals from the

aforementioned cohorts, in which it has been proposed jointly by WP1 and WP3 to verify the

feasibility of collecting and biobanking biological samples, which would allow for subsequent

analyses of pertinent biomarkers. The details of the strategy proposed for the collection of the

biological data (standard operating procedures for biological sample collection, transportation,

storage and biomarker testing) will be presented in the “biology protocol for a molecular

epidemiology study ” section 4.2. below.

Epidemiologist, biologists and dosimetrists first examined the possibilities to launch prospective pilot

studies in workers currently active, which would allow for a repeated collection of samples over time

in the same workers. This collection scheme would allow for an analysis of biomarkers (of exposure,

then of early biological effects) before the onset of any radiation-induced disease so that the kinetics

25

of both uranium exposure and biomarkers, as well as their associations, can be studied over time.

Starting from these criteria, several potential population subgroups from the aforementioned

European cohorts of uranium workers have been proposed jointly by WP1 and WP3:

uranium workers currently employed at an AREVA uranium conversion plant in Malvesi

(France), N~250.

former uranium workers now involved in decommissioning activities in Belgium and non-

exposed workers, N~40.

Uranium millers and miners currently employed at the Rozna mine (Czech Republic), N~100.

The feasibility of collecting biological materials in these subcohorts was investigated in detail (see

section 4.2.5.).

In addition, information was collected more briefly about possible opportunities to collect biological

samples in uranium workers in other settings (or to analyze biological samples already collected

there) in the future. This includes uranium workers employed in European as well as non-European

countries. The contacts established with invited experts have been very important for that purpose:

Preliminary contacts have been established with Polat Kazymbet and Meirat Batkin (Astana

Medical University, Kazakhstan) to examine the feasibility to access samples previously

collected in Kazakh workers, or to collect new samples. Some samples have already been

collected in uranium workers from the Stepnogorsk site, notably for the purpose of gene

expression analyses (Abilmazhinova et al., 2014; Kazymbet, 2014).

Contacts have been established with Andrey Karpov and Ravil M Takhauov (Seversk

Biophysical Institute, Russia) to examine the feasibility to analyze samples previously

collected in uranium workers in Seversk (Russia), or to collect new ones. A large biobank has

been set up there (Takhauov et al., 2015).

Contacts have been established with Friedo Zölzer (University of South Bohemia) who has

conducted a pilot study in uranium miners and millers at the Mydlovary site (Czech

Republic)(Zolzer et al., 2012b). These miners and millers have already been sampled twice,

with a 90 % participation rate for the follow up one year after initial sampling.

First contacts have been established by Nuvia Ltd to assess whether a feasibility assessment

could be possibly conducted in uranium workers the UK (Dounreay, and Urenco at

Capenhurst).

Lastly, the possibility to examine biological samples collected a long time after exposure was also

examined. It is of great interest to identify persistent biomarkers of exposure or examine biomarkers

of late biological effects such as a specific biological signature of radiation-induced disease (e.g.

uranium specific “fingerprints” in tumor tissue). A biobank including former uranium miners retired

from the Wismut company (Germany) has been set up at BfS (Gomolka et al., 2012). This biobank

may provide interesting material for such analyses.

26

4.2. Biological protocol for a molecular epidemiology study

The aim of the biological protocol is to provide a strategy as well as standardized operating

procedures (SOPs) and study instruments to perform relevant biological sampling, biomarker

measurements and analysis in a cohort of uranium exposed workers, in order to constitute an

international biobank.

The rationales for establishing such a biobank and integrating measurements of biomarkers in a

cohort study have been presented in section 1.6.2. Briefly, the long-term goal is to set up a large

international molecular epidemiology study that will provide new insight into the biological effects

associated with occupational exposure to uranium and help refine the estimates of related health

risk. Another potential side product of this research might be the identification or validation of new

biomarkers useful to the surveillance of workers occupationally exposed to uranium.

Setting up an international biobank including biological samples from workers is quite a novel and

complex task. It is anticipated that approval of stakeholders and ethics as well as data safety

committees will be possible only if a clear strategy is provided together with a strong management

system that ensures the data analysis. The biological protocol therefore defines the following aspects

of the proposed study:

A list of biomarkers of interest to study in relation with uranium exposure.

A related list of biospecimens that should be collected for the measurement of the selected biomarkers (as well as of other biomarkers which may be proposed in the future).

Standardized operating procedures (SOPs) for biospecimen collection, processing,

transportation, storage and for biomarker testing/measurement. Some SOPs for functional

tests were also proposed (e.g: in order to assess the cerebral or circulatory functions of

workers) (Gomolka et al., 2014).

A questionnaire to collect the information needed for an adequate interpretation of

biomarkers and appropriate statistical analysis

An information sheet for workers and related form of consent to participate to the study

In addition, since the successful measurement of most biomarkers require stringent

conditions of sampling, processing, storage and transportation of specific biospecimen

(which may widely differ according to the biomarkers which are intended to be measured in

them), it is necessary to propose, assess, and if necessary refine, a precise logistic strategy

allowing for the collection of several biospecimens in one or more pilot cohort(s). Another

mandatory step before moving to a large scale molecular epidemiology study would be to

conduct one or more pilot studies for field testing. Such pilot studies would notably be of

interest to assess the participation rate among workers, the appropriate completion of

questionnaires, the applicability of SOPs in field conditions on a real scale (e.g.: for several

workers per day), the quality of resulting samples for the envisaged biomarker analyses.

Whilst launching such pilot studies was clearly beyond the time frame of the CURE project it

27

was recognized that preliminary feasibility studies should be conducted in the groups of pilot

cohorts proposed jointly by WP1 and WP3, to assess the compatibility of the proposed

logistics protocol with field conditions.

All these aspects are detailed in the biological protocol below.

4.2.1. Proposed biomarkers

A list of biomarkers of interest to study the biological effects of uranium has been proposed. This list

has been established by taking into consideration a priori the two main types of approaches

currently used in biomarkers research:

Targeted approaches. Biomarkers that were considered as potentially informative based on

the knowledge of uranium action in humans or animals were identified. This includes

biomarkers that may reflect damage to organs/tissues/systems identified as established or

potential targets of uranium according to published biokinetic, experimental or

epidemiological studies. This list of biomarkers covers early as well as late biological effects

in the target organs/tissues/systems (lung, kidney, heart, bone, blood, vessels, red bone

marrow and central nervous system). Because no biomarker identified so far is specific to

uranium exposure or to its suspected health effects, a multi-marker approach has been

proposed for most targeted biological endpoints. Using a panel of relevant biomarkers will

allow for a better delineation of the suspected targeted effects. Multi-marker approaches

will also be useful to explore the interplay of different possible biological pathways (Pernot

et al., 2012).

Non-targeted approaches (such as OMIC techniques) are highly dimensional multi-marker

approaches by nature. They aim at the collective characterization and quantification of pools

of biological endpoints that translate into the structure, function, and dynamics of

organisms. Such approaches are potentially able to identify new biomarkers of exposure

(Grison et al., 2013) and to detect perturbations of biological pathways by uranium exposure,

which have not been suspected so far. This might in turn lead to the identification of new

target organs/tissues/systems of uranium exposure and related health effects that can be

identified by new related biomarkers. In addition, quantitative meta analyses of differentially

regulated molecular levels by toxic compounds - from DNA to RNA to proteins to metabolites

and their regulatory factors (e.g. miRNA, DNA methylation) – should allow for a better/in-

depth understanding of their systemic action and induced biological mechanisms. Such

methods should help to improve radiation risk assessment in the future.

The various biological endpoints covered by the proposed list of biomarkers are listed below. The

detailed list of proposed biomarkers is provided in the standardized operating procedures produced

by CURE WP3 (Gomolka et al., 2014):

Lung cancer and other lung damages, because of first entrance and deposition by uranium

dust inhalation in the lung, and of suggestive results from animal and epidemiological

studies.

28

Renal damages, because of uranium accumulation and biological data from animal and

epidemiological studies indicating effects of uranium on kidney

Bone damages, because of uranium accumulation and long term storage in this tissue

Cardio-vascular damages, because of both direct and indirect effect of uranium through

kidney impairment, of the established effect of particle inhalation on the cardio-vascular

system, and of suggestive results from an epidemiological study

Lympho-hematopoietic system, because of clearance of uranium by macrophages and

uranium deposition in bones and lymph nodes, and suggestive results from an

epidemiological study

Brain damage, since changes in cognitive functions have been observed in animal

experiments but no data are available in humans

DNA damages to evaluate the radiotoxic and chemotoxic effects of uranium on genetic

material, using cytogenetic biomarkers

Metabolism because of previous identification of urinary metabolomic signatures of uranium

exposure in rats

Other, non-specific OMIC biomarkers to develop new hypothesis like RNA profiles and whole

genome RNA sequencing in order to detect fusion proteins (leukemia), alternative splicing

and micro RNA, DNA sequencing to detect methylation.

This extensive list includes a large number of biomarkers (as detailed in the SOPs), which may need

to be adapted in the context of a proposed study depending on practical constraints (amount of

funding available, samples for which expectations of successful collection are good). It is open for the

addition of emerging biomarkers of interest, since the proper collection and biobanking of the

biospecimen will provide the opportunity for a multitude of analyses in the future.

The defined biomarkers do not have to be all investigated in a first step. Indeed the quality of the

collected and stored material must allow for their future investigation, as well as for the future

analyses of emerging biomarkers. The best strategy for the identification of certain biomarkers

would be to wait until sequencing strategies become much more elaborated and cheaper.

4.2.2. Biospecimen to be sampled

As part of the CURE project, it was recommended to collect the following biospecimen from study

participants in order to investigate the proposed biomarkers:

Blood

Urine

Sputum

Nasal swabs

Saliva

Again, ad hoc SOPs are provided for the purpose of sampling biospecimen in the best conditions for

later measurement of specific biomarkers (Gomolka et al., 2014). In addition, general considerations

are provided below regarding the collection of these different types of biospecimen:

29

4.2.2.1. Blood

Blood is an important biospecimen to collect because of the high number of biomarkers that can be

measured in it with high quality. It has been estimated by members of the CURE project that a

volume of blood of about 50 ml per donor is acceptable, from both ethical and physiological point of

views. Mentioning preceding studies sampling similar quantities may help convince occupational

physicians who are not familiar with molecular epidemiology studies. For instance, up to 80 ml per

donor has been collected from retired miners as part of a pilot study for the Wismut Biobank

(Gomolka et al., 2012). In the UK biobank for which 500.000 individuals have been sampled a total

volume of 45 ml of blood has been collected per person (Elliott and Peakman, 2008).

Blood must be collected after overnight fasting, using classical blood collection sets with BD

Vacutainer-type, safety-lock tubes, pre-coated with special preservatives, each destined for a specific

use and the measurement of different sets of biomarkers. A well-defined blood collection protocol,

sampling instructions and the number and type of the collected tubes have to be communicated

clearly to nurses in charge of blood collection. Nurses should be trained to follow the designed

protocol. It should be noted that some centrifugation steps have to be performed shortly after

sampling either on site or in a classical medical laboratory located near the sampling site.

Participating individuals have to be informed about the risks associated with blood sampling

(venipuncture) by the medical staff a priori.

Adequate conditions for the processing, transportation and storage of blood samples can be

summarized as follow. These ideal conditions for sampling might need to be adjusted according to

the site specific conditions:

Table 1. preferred conditions for blood sampling, processing and storage

Purpose Tube type Biological Investigation

On-site Processing

Transportation Storage

DNA isolation K2-EDTA tubes DNA damage none room temperature

-20°C

Lymphocytes isolations Lithium–Heparin tubes

translocations none room temperature

Rapid isolation of lymphocytes (max 2 days)

Plasma collection after 12h of fasting

Lithium–Heparin tubes

metabolomics, cardiovascular or brain markers

plasma collection and freezing

dry ice -80°C

RNA isolation *PAXgene tubes genomics none room temperature

-20°C (24h) then -80°C

Serum collection after 12h of fasting

SST tubes cardiovascular and brain biomarkers

serum collection and freezing

dry ice -80°C

Plasma collection after 12h of fasting

K2-EDTA tubes kidney, cardiovascular and brain biomarkers

plasma collection and freezing

dry ice -80 °C

30

4.2.2.2. Urine

Urine sampling is non-invasive and provides a biological sample that is of high interest as a matrix for

measuring kidney function biomarkers and metabolites. Two different protocols for urine collection

are proposed, each for a specific purpose, and are briefly summarized below:

A 24-hour collection time, used for studying of kidney biomarkers: The first morning urine sample

should be discarded, and then all urine should be collected over a 24 hour period until the first urine

sample of the following morning (see SOPs in (Gomolka et al., 2014)).

Following this collection, a second 2-hour urine sample should be obtained for metabolomics studies

and the analysis of additional kidney biomarkers. In order to control variation in metabolite

composition of the collected samples due to influencing factors (e.g. composition of meals before

sampling), a detailed list of recommendations (diet and physical activity restrictions) will be provided

before 2-hour urine sampling (see SOPs in (Gomolka et al., 2014)).

4.2.2.3. Sputum

The mucoid material that is expelled from the respiratory passages or the “breathing tubes” is called

sputum. Sputum is a highly interesting biological sample widely used in lung cancer studies, mainly

for the detection of specific gene methylation patterns as pre-diagnostic markers. It is also of interest

to investigate lung damages other than lung cancer (e.g., via measurement of the CC16 protein,

which is potentially indicative of obstructive diseases). In addition, sputum samples potentially of

interest for the measurement of uranium, as a potential source of information on the solubility of

uranium particles encountered in specific settings.

Again two different protocols can be envisaged. It is possible to collect either induced or

spontaneous sputum:

The collection of induced sputum is a quite complex procedure, as summarized below:

Preliminary spirometer test in order to evaluate breathing capacity of the participant.

Inhalation of a short acting β2-agonist and a succession of saline solutions of various

concentrations

Collection of induced sputum

Processing within 2 hours for cell isolation and conservation.

This collection requests the presence of a nurse and a private room for the participant. Its feasibility

in occupational settings would clearly need to be assessed via a pilot study in field conditions.

Alternatively, a less constraining procedure is to ask workers to collect morning spontaneous sputum

(Belinsky et al., 2005) at home over three days. A specimen can be obtained by coughing up sputum

from deep in the lungs and placing it in a bottle containing fixative. This fixative preserves the cells

until they can be prepared for evaluation in the laboratory. No freezing is necessary and samples may

even be sent by regular mail. On the logistics point of view, the coupling of such a collection with 24h

urine collection, which is also partly conducted at home, may be considered.

31

4.2.2.4. Nasal swabs

During inhalation, the nasal epithelium is directly exposed to environmental pollutants, including

airborne uranium particles. In addition the nasal epithelium is directly connected to, and may reflect

what happens in, the lung. Nasal swabs are of minimal cost and might provide important information

about exposure to uranium or biomarkers of early or late biological effects in the lung. For instance,

they can be used for the purpose DNA methylation assays. The procedure is not painful, but slightly

invasive. It requires gesture training from the nurse. The acceptability of such sampling appears

worth testing as part of pilot studies in workers.

4.2.2.5. Saliva

The usefulness of saliva samples for biomarker studies in radiation research (Pernot et al., 2014) or

to study the effects of internal contamination by uranium has not been extensively evaluated so far.

However, saliva is of interest as it contains a wide range of molecules, including proteins, peptides,

DNAs, hormones, metabolites and RNAs - including non-protein coding RNAs (ncRNAs) like

microRNAs (miRNAs). The use of saliva for genetic analyses has grown dramatically in recent years in

molecular epidemiology studies and recent characterizations of the salivary proteome and

transcriptome have highlighted the diagnostic potential of saliva. Saliva contains clinically

discriminatory protein and RNA biomarkers of oral cancer for example. Although some confounding

factors have to be taken into account (tooth brushing, oral disease, microbiome in the mouth,

circadian rhythm …) and that blood generally has a bigger potential, the collection of saliva is non-

invasive and easy to perform. A good level of acceptance can therefore be expected from

participants. It can be collected in large volumes in appropriate buffers and stored at room

temperature for subsequent genomic DNA and RNA isolation. Saliva might also be a useful

biospecimen for uranium measurements and dosimetric purposes, with potential to reflect lung and

oropharynx cavity contaminations. Although the relative usefulness of saliva samples versus other

samples for the purpose of studying the effects of uranium exposure cannot be guaranteed to date,

it would be of interest to test their potential as part of a pilot study in uranium workers.

4.2.2.6. Further considerations on biological samples

Again, the collection of biological samples proposed above corresponds to an ideal sampling plan. In

some settings, the collection of certain biological samples may be refused for practical reasons (e.g.

important changes needed in the schedule of the medical check-ups to integrate their collection, lack

of availability of a dedicated room, etc) or limited in volume (typically, competition between the

volume of blood collected for routine medical surveillance and the volume needed for the study).

Other adaptations may be needed according to field conditions. For instance, practical constraints

regarding temperature conditions and equipment or time to sample processing might not exactly fit

the proposed SOPs in some settings. Unless these conditions can be changed, the robustness of the

SOPs to the field conditions should be quickly tested on a few samples (e.g.; replication of conditions

in a lab) to make sure that the modifications of conditions/procedures do not impair the quality of

biological samples for future analyses.

32

Several biomarkers can be measured in different kinds of biological samples (e.g., CC16 marker for

lung damage can be measured in sputum, serum and plasma). This may represent alternative

solutions if one type of biological sample cannot be collected. Alternatively, measuring the same

biomarker in two different types of biospecimen (especially, more or less invasive or costly) may

prove of interest for the purpose of comparison and validation studies. Applications of such

comparisons would apply to workers studies and possibly other situations (emergency, accidents)

where time and resources are more limited for the collection of certain specimen (typically, blood or

sputum) or not easily acceptable for other reasons (e.g., studies in children).

4.2.3. Questionnaire

A questionnaire to be completed by all participants in the study at the time of biological sampling

was produced by CURE partners and is provided in (Gomolka et al., 2014). In complement to the

retrospective information available in medical records, it will allow confounding factors to be

identified and taken into account in the statistical analysis of biological data.

The questionnaire should ideally be filled-in by the worker at the center (on-site, before the blood

sampling) and if possible verified at least in part by the physician (or by an associate) during the

medical exam.

Specific and detailed questions are included in the proposed questionnaire, related to:

Demographic characteristics

Employment history

Socioeconomic status

General health status

Cardiovascular diseases

Cancer

Pulmonary diseases and respiratory symptoms

Kidney impairment

Thyroid impairment

Autoimmune disease, diabetes

Medications (anti-coagulant, anti-inflammatory, statins, pain killers)

Dietary supplements (vitamins, calcium, etc), ,

Hormones

Transplantations and transfusions

Infections

Reporting of radiation exposure for medical reasons including nuclear medicine,

interventional radiography, catheter exam, radiography, radiotherapy

Chemotherapy.

Alcohol consumption

Smoking habits

Physical activity and sleeping habits

33

Again, the questionnaire produced as part of the current protocol is extensive. It may need to be

adapted (e.g., shortened) according to the list of biomarkers selected for investigation, depending on

the amount of funding available and to other practical constraints.

4.2.4. Information sheet and consent form

For the purpose of requesting and collecting an informed consent from the workers willing to

participate to the study (i.e., to fill in questionnaire and provide researchers with their biological

samples) and keeping track of it, two study instruments were produced:

An information sheet describing the aim and main characteristics of the research project, as

well as the participant’s rights (Gomolka et al., 2014).

An informed consent form (Certificate of Consent) to be signed by the participants and

conserved by the investigators during the full duration of the study and beyond (Gomolka et

al., 2014)

4.2.5. Logistic strategy for the collection of biospecimens in pilot cohorts

The aim here was to propose a working logistic strategy to be implemented for the collection,

processing, transportation and storage of several biospecimens according to the defined SOPs

(Gomolka et al., 2014), but also to inform workers about the study, obtain and document their

informed consent. A time also needs to be allocated to allow the workers to fill out the questionnaire

and undergo complementary examinations, such as proposed functional tests for brain and

circulatory functions (Gomolka et al., 2014).

The proposed logistic strategy needs to:

be compatible with field conditions (timing of the medical check-ups, usual practices and

specific considerations of occupational medicine staffs, resources locally available)

allow for the collection of biospecimens useful to measure a wide array of biomarkers for

which processing or storage conditions could markedly differ, in a single logistic frame

allowing for a cost-efficient approach.

Logistic strategies were therefore developed for the pilot cohorts proposed jointly by WP1 and WP3.

34

4.2.5.1. Proposed French pilot cohort

A feasibility study was conducted at the AREVA uranium conversion plant in Malvesi (France) to set

up a logistics strategy integrating the collection, processing, transportation and long-term storage of

biospecimen (biobanking) that would allow for the measurement a panel of biomarkers in which

CURE partners expressed immediate interest:

metabolomics (IRSN)

gene expression (SCK•CEN, PHE)

kidney function and damage markers (IRSN)

lung damage markers (CREAL)

central nervous system markers (IRSN)

cytogenetics (BfS)

cardiovascular diseases markers (IRSN)

The main features of this feasibility study are presented below.

Context and field conditions

The Malvesi plant in Narbonne (southern France) has performed purification of natural uranium and

its conversion to UF4, since the beginning of its operations in 1958. This plant was fully integrated

into the AREVA group in year 2013. At present, 250 active workers are holding a stable professional

position there. Among them, about 100 perform shift work and are monitored for uranium exposure.

Once a year, these workers undergo a check-up by the occupational medicine service, including

blood and 2h-urine sampling for the purpose of routine medical surveillance. These samplings have

been conducted in the morning (from 8 am) after overnight fasting, every day from Tuesday to

Friday. Three tubes of blood per worker have been routinely collected (EDTA (3.5 ml), serum with

clot activator (6 ml) and Sodium Fluoride and Potassium oxalate (3.5 ml)) and sent to a subcontractor

medical laboratory (a 15-minute drive away from Malvesi) for analysis on the day of sampling, along

with the 2h-urine samples. Urine and blood samples are usually analysed before 2pm, and stay at

room temperature between sampling and analyses. According to the medical laboratory, such

temperature conditions could not be changed for the 3 tubes of blood routinely collected.

In addition, 24h-urine samples are collected once a year during specific campaigns (at times distinct

from those of medical check-ups), for the purpose of uranium measurement. Extra samplings are

also conducted in case of suspected or confirmed contamination. Uranium measurements in urine

are conducted in a separate laboratory in Marcoule (France).

Collection of biological samples proposed to the medical staff

The following collection of biological samples for a pilot study was proposed to the occupational

physician and nurses:

The collection of an aliquot of 2h urine samples (10 ml) was accepted

The collection of an aliquot of 24h urine samples (10 ml) was accepted

The collection of saliva samples was accepted

The collection of spontaneous sputum at home and of nasal swabs was also proposed but

refused by the occupational physician

35

The collection of any unused content of the blood tubes routinely sent to the laboratory (1 to

1.5 ml from EDTA tube, plus 1 to 1.5 ml from the serum separating tube) for the purpose of

the pilot study was accepted

The collection of four more tubes of blood per worker for the purpose of the study was

accepted:

o 2 heparin (10 ml) tubes (1 for metabolomics and plasma markers, one for

cytogenetics)

o 1 EDTA (10 ml) tube (for plasma markers)

o 1 PaxGene (2.5 ml) tube (for RNA)

In total the sum of the volumes collected a) for the purpose of the routine medical surveillance and

b) specifically for the purpose of the pilot study per worker would be 45.5 ml, divided into 7 tubes.

Logistic strategy

The occupational physician and nurses at the Malvesi site clearly asked for logistic solutions that

would keep their routine medical surveillance activities unchanged. They also expressed a preference

for the processing of samples to be conducted outside of the medical building because of a lack of

appropriate rooms available on site.

These requests had to be taken into account in the procedures, notably since the acceptance and

support from the occupational physician and nurses is important to motivate workers to participate

to the study.

It was jointly decided with the medical staff:

to hire a part time subcontractor Clinical Research Associate (CRA) in order to help with

several tasks related to the study

to mandate the subcontractor medical laboratory that was conducting the routine

surveillance analyses to process the additional samples and to ensure their temporary

storage (before shipping the samples to a biobank)

to organise the long-term storage of biological samples in a standardised biobank belonging

to the European BBMRI infrastructure (bbmri-eric.eu). Biobanks which would agree to store

the proposed samples were identified with help from the infrastructure BIOBANQUES

(http://www.biobanques.eu).

On the basis of the above field conditions and requests from the medical staff, the following logistic

scheme was defined:

The nurses would sent convocations to workers as usual (email and letters), along with the

information sheet about the study and with polite requests, for workers willing to participate, to

follow the food and physical activity restrictions for metabolomics on the night before sampling.

On the day before sampling, the CRA would label blood tubes, urine containers, saliva collection kits

and cryotubes with anonymized identifiers for the subject and explicit codes for the specific

sample/aliquot, according to the classical recommendations of biobanks (Henny et al., 2013).

36

On the day of sampling, the CRA would approach workers along with the nurses to remind them

about the aim of the study with help from the information sheet, and provide additional information

if needed (e.g., reply to any complementary question about the study). No incentive would be

proposed to workers since this is forbidden by French regulations.

The CRA would then collect the informed consent of workers using the appropriate form.

The workers would then provide 2h-urine samples as usual to the nurse, and the CRA would collect

one aliquot of 10 ml from this sample for the purpose of the study.

Next, the nurse would conduct the blood sampling as usual, but for the 7 tubes instead of the usual

4, with assistance from the CRA who would provide her with the appropriate pre-labelled blood

collection tubes, and handle them immediately after sampling. Two tubes (EDTA+heparin) collected

for the purpose of measuring plasma markers and metabolomics would be transferred immediately

into an electric cool box at 4°C, along with the 2h urine samples, for shipping to the subcontractor

laboratory. Other collected tubes (heparin tube for cytogenetics, PAXgene tube for RNA analyses,

serum separating and EDTA tubes usually collected for routine medical surveillance) would be kept at

ambient temperature before shipping.

The CRA would then invite the workers to enter an isolated room, provide them with a saliva

collection kit and related instructions. the CRA would then collect the saliva sample and provide the

workers with a snack and with the questionnaire. After completion of the questionnaire, the CRA

would briefly check that it has been adequately completed, and administrate the cerebral function

test (Mini Mental State Evaluation (Folstein et al., 1975.)) and vascular reactivity functional tests (see

(Gomolka et al., 2014)).

By noon, all collected samples (except the heparin tube for cytogenetics) would be shipped to the

subcontractor laboratory by the specialized transporter usually employed for that purpose, either in

the cool box or at ambient temperature, depending on the requirement for each specific sample. The

cryotubes previously labelled with anonymised identifiers by the CRA would also be sent the same

way to the subcontractor laboratory.

Immediately at reception, the laboratory would perform the following tasks:

centrifuge the EDTA and heparin tubes collected for the purpose of measuring plasma

markers and metabolomics and aliquot the plasma in the pre-labelled cryotubes.

centrifuge the serum separating tube collected for the purpose of regular occupational

surveillance, measure usual surveillance parameters, and aliquot the remaining serum in the

pre-labelled cryotubes.

analyse parameters in the whole blood of the EDTA tube collected for the purpose of regular

occupational surveillance, then mix the remaining whole blood (about 500 µl) with 1.3ml of

RNALater to get genomic DNA and aliquot it in the pre-labelled cryotubes

aliquot the 10 ml 2h-urine aliquot in the pre-labelled cryotubes

incubate the saliva samples and aliquot them in the pre-labelled cryotubes

All aliquots (pre-labelled by the CRA) would then be stored in a -80°C freezer provided to the

subcontractor analysis laboratory, along with the PAXgene tube.

37

Once per month, the aliquots would be shipped on dry ice by a specialized transporter from the

subcontractor laboratory to a biobank affiliated to the BBMRI network. The maximum duration for

local storage of aliquots in the laboratory would therefore be one month. In the biobank, the

aforementioned aliquots and the PAXgene tube would be stored at -80°C until shipping to specialized

centers (laboratories/platform) for biomarker measurements. RNA would need to be extracted from

the PAXgene tube within two years of r collection.

The heparin tube collected for cytogenetics would follow a different pathway after sampling, since it

would be shipped directly from the Malvesi site to the local biobank at ambient temperature by a

specialized transporter. Once in the biobank, the heparin tube would undergo a procedure of

lymphocytes extraction (Ficoll gradient, see (Gomolka et al., 2014)). The extracted lymphocytes

would remain stored in the biobank, cryopreserved in liquid nitrogen until shipping to specialized

centers (Gomolka et al., 2014) for biomarker measurements.

Because biological samples are collected once a year for each worker, the duration of the first round

of sample collection, corresponding to the length of the pilot study, would be one year in order to

cover the entire target population of workers.

It was agreed that to avoid batch variations the best option for the analyses of biomarkers would be

to analyse samples collected for all workers in a row on one platform (per biomarker). Therefore it

would be necessary to wait at least until the end of the first round of sample collection, which would

be one year after the start of the pilot study, before shipping samples to specialized centers for

biomarker measurements.

Conclusion

It is technically feasible to collect biological samples for studying the biological effects of uranium

exposure in the Malvesi conversion plant, although the occupational physician disagreed on the

principle of spontaneous sputum collection and nasal swabs. A workable and realistic logistic strategy

was developed, that would need to be tested by launching a pilot study.

After a presentation of the study project to the Health and Safety committee (Comité d’Hygiène, de

Sécurité et des Conditions de Travail) of the Malvesi plant on October 16, 2014, the director, the

occupational physician and the workers representative committee of the Malvesi plant have agreed

to the principle of the pilot study.

However for this pilot study to be launched the financial support of the AREVA Health

Direction/management is necessary.

4.2.5.2. Proposed Belgian pilot cohort

The technical feasibility of a similar scheme was also evaluated for a pilot cohort of 40 former FBFC

workers (20 formerly exposed, and 20 non exposed to uranium) monitored by SCK•CEN occupational

medicine.

38

Preliminary investigations have demonstrated the technical feasibility of a collection of biological

material of interest to study the biological effects of uranium exposure in these workers, according

to a logistic strategy similar to the one presented above for the Malvesi plant.

However, workers formerly employed by FBFC are now affiliated to AREVA. Therefore this pilot study

can only be launched provided that AREVA Health Direction/management supports it.

4.2.5.3. Proposed Czech pilot cohort

Context and field conditions

A feasibility study was also conducted in the Czech Republic at the Rozna uranium mine and milling

plan, which has been operating since 1958. About 440 miners and millers are currently employed

there and conduct a large number of activities including extraction, drying, crushing and chemical

processing of uranium, and others. About 30 workers are employed at the milling plant, which is

open during 5 month per year. Medical check-ups in Rozna are conducted once a year for each

worker, on Tuesdays, from 6 a.m. to noon. Some miners perform shift work, but those showing up

for the medical check-ups on Tuesdays will have worked only in the morning the day before (i.e.: on

Monday).

A research project supported by the R&D program of the State Office for Nuclear Safety of the Czech

Republic and by DoReMi is ongoing at the Rozna mine (IntEmitUM: Internal Emitters in Uranium

Miners, DoReMi task 5.5.1). A component of this project will include a comparison of the dosimetry

of current uranium miners computed from personal dosimeters (ALGADE), with the dosimetry

computed from measurement of uranium in the urine of these same miners. For the purpose of this

research, spot urine samples from uranium miners have been collected during regular medical check-

ups, however they have not been stored under conditions that would be appropriate for later

analyses of metabolomics or kidney markers.

Collection of biological samples proposed to the medical staff

Within the framework of CURE, the following collection of biological samples for a pilot study was

proposed to the occupational physician and nurses:

The continued collection of further aliquots of 2h urine samples (10 ml) for the purpose of

metabolomics and kidney markers analyses has been accepted

The collection of saliva samples has been accepted

The collection of two tubes of EDTA blood (2*8 ml) on top of the amount already collected

for the purpose of the routine medical surveillance has been accepted.

It was proposed to invite the workers to a medical center in Prague to organize

complementary examinations such as magnetic resonance imagining (for the study of central

nervous system), measurement of intima media thickness (for the study of the

cardiovascular system) and to collect induced sputum, but the occupational physician

refused this option.

Logistic strategy

According to a general strategy similar to the one developed in Malvesi, it has been proposed to

collect the informed consent, the biological samples and administrate the questionnaire during the

39

annual medical check-ups of workers in Rozna. In The Czech Republic, incentives can be proposed to

workers and medical staff. This possibility would be used to maximize the participation rate. An

incentive of about 20 euros per workers has been proposed.

The samples collected on each Tuesday would be shipped to a laboratory in Bystřice nad

Pernštejnem, a 10 minutes drive away from Rozna. Two shipping per morning would be possible.

Immediately at their reception in the laboratory, the samples would be processed and kept frozen in

a -80°C freezer for several weeks. The aliquots would then be shipped on dry ice by a specialized

transporter from the subcontractor laboratory to BfS for biobanking. From there, samples would be

dispatched toward specialized centres for biomarker measurements under the responsibility of CURE

partners.

Collection of the biological material (for 100 miners and millers) would be expected to take 6

months.

A translations of WP3 reference documents (Gomolka et al., 2014) in the Czech language will be

required.

Conclusion

This preliminary study confirms that it is technically feasible to collect biological samples for studying

the biological effects of uranium exposure at the Rozna mill and mine.

40

4.2.5.4. German Uranium Miners Biobank

Another strategy to access biospecimens of interest for molecular epidemiology studies on the

biological effects or uranium exposure is to use readily established biobanks of individuals exposed

to uranium, such as uranium miners.

In 2009, the BfS initiated a biobanking strategy from former Wismut Miners. Supported by the

German Social Accident Insurance (DGUV), the Institute for Prevention and Occupational Medicine of

the German Social Accident (IPA) in Bochum, the Federal Institute for Occupational Safety and Health

(BAuA) and the Institute of Epidemiology of Helmholtz Center Munich, various biological materials

from healthy controls and lung cancer cases of former Wismut miners and their children have been

collected and stored in the BfS biobank (Gomolka et al., 2012).

Standard operating procedures have been developed to establish a biobank based on blood samples

of 65 ml to 80 ml in a preceding pilot study. 442 volunteers have been recruited for sample

collection. Sample collection and shipment was performed as simply as possible and the quality of

isolated lymphocytes, DNA and RNA, as well as protein degradation from plasma samples was tested.

In most cases high quality material was stored and is suitable for various OMIC techniques such as

DNA and RNA whole genome sequencing, methylation analysis, microarray analyses of RNA and

miRNA expression. The following biospecimens were stored now from healthy miners and partially

from children of former miners who died with lung cancer: in liquid nitrogen stored lymphocytes

(from Heparin and EDTA conserved blood samples), DNA and RNA as well as Paxgene samples ready

for further RNA isolation and plasma samples from EDTA tubes suitable for the analyses of proteins

such as cytokines as possible for the analyses of metabolites. 250 DNA and 50 RNA samples are

available from tumor and normal tissue derived from the pathological archive.

In addition biomarker research has been started and data are available and will be accessible in the

European STORE data bank for meta-analyses of various molecular levels.

These data comprise microarray analyses (Affymetrix U219 Microarray) in 200 miners

(100High/100low exposed according to red bone marrow exposure), RNAseq analyses in 20 miners

(10High/10low exposed according to red bone marrow exposure), miRNA analyses on a self-spotted

microarray (703 human miRNAs, 34 non validated miRNAs and 12 controls) in 60 miners

(30High/30low exposed according to lung exposure), SNP analyses of the ATM gene (15 SNPs) in all

414 samples, specific miRNAs in all 414 samples (miRNA-26a und miRNA-26b), DNA methylation of

ataxia telangiectasia mutated (ATM) promoter/ long interspersed nuclear element (LINEI) in all 414

samples, ATM expression versus GAPDH expression in all 414 samples. Oncoarray-Chip data will be

generated for all samples in the next year.

41

4.3. Dosimetric protocol

4.3.1. Introduction

4.3.1.1. Objectives

This section provides an overview of a protocol for the reconstruction of annual received doses

within the proposed epidemiological and molecular epidemiology studies in uranium miners and

workers cohorts. More details are available within the two deliverables (D2.1. and D2.2.) of the

dosimetry workpackage WP2 of the CURE project (Blanchardon et al., 2014a; Blanchardon et al.,

2014b).

To allow for the study of the epidemiological and biological endpoints of interest defined in the

previous sections, annual absorbed doses (from the first year of radiation work to the end of follow-

up) resulting from uranium exposure will be calculated for the following organs and tissues: regions

of the lung (alveolar-interstitial, bronchial, and bronchiolar), red bone marrow, liver, kidney,

stomach, small intestine, colon, upper airways (mouth and nose), endosteum (bone surfaces), heart,

kidney, lymph nodes and brain of each individual worker. Two sets of weighted annual lung doses

will be provided based either on regional mass weighting or assuming equal detriment for each lung

region.

The separate contributions due to the exposure to other radiation sources than uranium will also be

provided where enough information is available to do so. Doses resulting from intakes of other

radionuclides than uranium (e.g. radon and its progeny, long-lived radionuclides in uranium ore,

plutonium, polonium, fission and activation products) will be calculated. Also the contributions

arising from the alpha radiation alone, as compared to the sum of alpha, beta and gamma radiation,

will be provided. The values of the total absorbed dose to specific organs will also include the

additional contribution of the external gamma radiation. The mean annual mass of uranium in the

lung, kidney and brain will be calculated to support investigation of its chemical toxicity, aside from

potential radiological toxicity.

4.3.1.2. Differences between uranium miners, millers and workers in dose assessment

procedures

For uranium miners and millers, the intake of radionuclides is usually determined from the

measurement and record of long-lived radionuclides, radon gas and radon progeny in the ambient

air. The size distribution of radioactive particles, the breathing rate and the duration of exposure are

significant exposure parameters that must be taken into account to determine the intake. The intake

is translated into the annual dose absorbed by radiosensitive tissues of the body through the

application of biokinetic and dosimetric models subject to the particles size and solubility. Alongside

uranium, the miners are exposed to other long-lived radionuclides in the uranium ore and, as

mentioned above, to radon gas and its short-lived progeny and external gamma exposure. Compared

with uranium miners, the millers have low radon exposures but still potentially high uranium intakes.

The milling operations and the related information from German and French mills were reviewed to

define specific parameter values for millers’ exposure. Similar dose assessment procedures as for

miners should be applied to millers.

42

Exposure of other workers involved in later stages of the nuclear fuel cycle is mainly to uranium

compounds and it is monitored by individual bioassay measurements (mainly in urine, but also in

faeces, whole-body or lungs). Occupational intakes are mostly by the inhalation route and are

estimated from the bioassay measurement results using a biokinetic model and additional

information on the exposure scenario including the type (acute/chronic) and time of intake as well as

the physicochemical form of the radionuclide. The estimate of intake is then translated into dose by

a dosimetric model.

In summary, the procedure of dose assessment for miners, millers and other uranium exposed

workers is therefore significantly different, although it makes use of the same biokinetic and

dosimetric models, which will be presented first (4.3.2). Subsequently, specific indications for other

steps of the dose reconstruction (parameters of exposure, 4.3.3; monitoring data, 4.3.4, and

assessment procedure 4.3.5) will be presented separately for miners and millers on the one hand,

and for workers on the other hand.

4.3.2. Biokinetic and dosimetric models

The reference biokinetic models have been developed mainly by the International Commission on

Radiological Protection (ICRP).

4.3.2.1. Routes of entry.

The calculations will be carried out by implementing the human respiratory tract model

(HRTM)(ICRP, 1994) revised in the Occupational Intakes of Radionuclides documents (ICRP, 2015a)

for inhalation intake, the biokinetic model for radionuclide contaminated wounds (NCRP, 2006) for

wound intake and the human alimentary tract model (HATM) (ICRP, 2006; ICRP, 2015a), for ingestion

intake.

The complex chemistry of uranium in the nuclear fuel cycle means that there is potential for

exposure to a number of compounds. These uranium compounds have very different solubilities in

biological fluids. This solubility governs the extent and the kinetics of absorption from lungs to blood,

leading to different retention and excretion kinetics. Knowledge and/or assumptions about the

chemical form of incorporated uranium is therefore important to the correct interpretation of

bioassay measurements and can also produce very large differences in estimated lung doses.

However the knowledge of the chemical forms of uranium to which a specific worker is exposed is

often lacking, creating a major source of uncertainty.

The workers exposure to uranium is monitored mainly by urine bioassay, which is directly connected

to the blood content and thus to the systemic organs activity, independently of lung solubility. On

the contrary, the relation between activity in lung and in blood completely depends on the solubility

of uranium. Uncertainty on lung solubility therefore generates significant uncertainty on intake

estimate, moderate uncertainty on systemic organ doses and strong uncertainty on the lung dose.

Lung solubility of uranium compounds has been identified as one the most important source of

uncertainties for the estimation of lung doses in uranium workers. These aspects, as well as

proposed strategies for progress, have been discussed by the UWG and will be developed in the

uncertainties section below (4.5.).

4.3.2.2. Systemic biokinetic models

43

The systemic biokinetic models for uranium (ICRP, 1995a; ICRP, 2015b) thorium, radon gas, actinium,

protactinium, polonium, lead, bismuth, and radium described in the ICRP Occupational Intakes of

Radionuclides publications (ICRP, 2015a; ICRP, 2015b; ICRP, 2015c; ICRP, 2015d) will be

implemented.

In the ICRP systemic model for uranium (ICRP, 1995a; ICRP, 2015b), following uptake to blood, about

75% of uranium is excreted in urine. About 15% is deposited on bone surfaces but only a small

amount is retained long term. The remainder is transferred to liver, red blood cells and other soft

tissues, while limited faecal excretion takes place. The decay products formed within the body

behave with their own specific biokinetic models; so-called independent kinetics.

The organ doses arising from the inhalation of radon gas alone will be evaluated with a biokinetic

model with parameters based on a blood flow model and specific tissue-to-blood partition

coefficients for radon gas (Leggett et al., 2013). Unlike the inhalation of radon progeny, the absorbed

dose to the lung arising from the inhalation of radon gas is independent of the breathing rate. It is

assumed that the gas rapidly saturates the lung and the dose mainly arises from the decays occurring

in the air within the lung. The absorbed dose to each region of the lung will be calculated assuming

that the activity in the volume of the gas within the airway can be replaced by the same activity,

uniformly deposited on the surface. Because of its relatively long half-life (3.8 d), radon gas is

susceptible to make a more significant contribution to systemic organ doses after absorption to

blood than its short-lived (<30 min) progeny which decays essentially in the lung.

4.3.2.3. Dosimetric models for internal exposure

The nature, yield and energy of radiation emitted during nuclear transformations will be considered

according to ICRP publication 107 (ICRP, 2008). The human anatomy will be represented by the

reference computational phantoms of ICRP publication 110 (ICRP, 2009). The resulting deposition of

energy in radiosensitive tissues will be calculated using the values of specific absorbed fractions

provided by the ICRP (ICRP, 2015e).

4.3.2.4. External dosimetry

Protection quantities such as effective dose defined by ICRP (ICRP, 2007) are not directly measurable.

However, conversion coefficients which relate protection quantities to physical quantities, such as air

kerma free-in-air, can be calculated using radiation transport codes and appropriate computational

models (phantoms). The ICRU (ICRU, 1993) defined operational quantities for area and individual

monitoring. These measurable quantities such as ambient dose equivalent, H*(d) and personal dose

equivalent, Hp(d) were designed to provide an estimate of protection quantities. Correction factors

proposed by (Marsh et al., 2009) will be applied to the gamma dose values given in the

epidemiological databases for uranium miners and workers in order to obtain an estimate of organ

absorbed dose DT.

4.3.3. Parameters of exposure

4.3.3.1. Miners and millers

Aerosol parameter values for uranium ore dust

44

An activity median aerodynamic diameter (AMAD) of 7 m was chosen to represent an aerosol of

long-lived radionuclides (LLR) in uranium ore dust for underground miners. An AMAD of 7 µm was

also chosen to represent aerosols of uranium in ore dust and in yellowcake for millers. For surface

miners an AMAD of 10 m was selected for an opencast mine. The HRTM defaults for geometric

standard deviation (σg) (2.5), density (3 g cm-3) and shape factor (1.5) will be assumed (Marsh et al.,

2008).

Some authors studied the solubility characteristics of the LLR in uranium ore dust by measuring the

undissolved fraction of the nuclides in simulated lung fluid (Bečková and Malátová, 2008; Duport et

al., 1991). Their results were used to derive absorption parameters (Marsh et al., 2009) for miners.

The values assumed for fA, the fraction of activity absorbed to blood from the small intestine, are the

ICRP Publication 68 (ICRP, 1995b) default values given for each element.

For millers, the derivation of absorption parameter values for different milling operations is

discussed in CURE deliverable D2.2 (Blanchardon et al., 2014a). As a result, the parameter values of

Table 2 are proposed.

Table 2: Uranium absorption parameter values for millers.

Processed material fr sr (d-1) ss (d

-1) fA

Uranium ore 0.2 0.8 1.4E-3 0.004

Non-calcined samples 0.8 1 5.0E-3 0.016

Calcined samples 0.3 3 5.0E-3 0.006

Aerosol parameter values for radon progeny

The radon progeny aerosol in the atmosphere is created in two steps. After decay of the radon gas,

the freshly formed radionuclides (218Po, 214Pb, 214Bi) react rapidly (< 1 s) with trace gases and vapours

and grow by cluster formation to form particles around 1 nm. These are referred to as unattached

particles. The unattached particles may also attach to existing aerosol particles in the atmosphere

within 1 – 100 s forming the so-called attached particles. Typically, the activity size distribution of the

attached particles in a mine can be described by a lognormal distribution with an AMAD between

100 nm and 400 nm. The size of the unattached progeny is assumed to remain constant in the lung.

However, some of the ambient aerosols are unstable in saturated air (i.e. hygroscopic) and are

assumed to grow instantaneously on inhalation by a given factor.

The activity deposited in each region of the lung notably depends on the activity size distribution of

the aerosol, let aside physiological parameters such as breathing rate that will be discussed below.

Representative aerosol parameter values were chosen for the different exposure scenarios after

reviewing the literature (Marsh et al., 2008) and are provided in (Blanchardon et al., 2014a).

Classification of miners

The individual miners have been classified according to their job (hewer, miller or other - if the job

type is not known, then a default job type will be assumed (Marsh et al., 2009)), to the condition of

45

work regarding drilling (wet or dry) and to characteristics of the mine they worked in: geographic

location, nature (underground or opencast), ventilation (poor, medium or good), presence of diesel

engines (yes or no) (Marsh et al. 2008). Three levels of physical activity have also been defined for

miners: low, medium and high. The average breathing rates assumed for low, medium and high

levels of physical activity are 1.0 m-3 h-1, 1.2 m-3 h-1 and 1.4 m-3 h-1 respectively. If the physical activity

is not specified then a medium level of physical activity will be assumed.

4.3.3.2. Uranium workers

Pulmonary absorption and particle size

Uranium workers included in this epidemiological study have been involved in various processes

including: enrichment of uranium, fuel fabrication, treatment of spent fuel and mixed oxide fuels

(MOX) preparation, uranium metal machining, and uranium chemistry research. During all these

operations, uranium is handled in different chemical forms: ammonium diuranate (ADU), yellow cake

(ADU containing impurities produced by uranium mills), chelated with tributyl phosphate (U-TBP),

UO3, UF4, UF6, UO2 and U3O8. In addition, uranium is mechanically modified to satisfy the quality

criteria of nuclear fuel. Therefore, workers have been exposed to uranium under very different

chemical forms and physical properties such as particle size and density.

In the absence of direct measurements, ICRP recommends reference values for AMAD of 5 µm for

workers, with a σg equal to 2.5 (ICRP, 1994; ICRP, 2015a). There is no, or very little, information

available for the AMAD from measurements of aerosols for the specific facilities of interest in this

study and over the time periods of interest. However, it is known that particle size parameters have a

relatively small effect on assessed intakes and doses, over the range of values typically found in

occupational environments (Riddell, 2005). Therefore the reference ICRP values for AMAD and σg will

be applied by default to the uranium workers in this study.

The ICRP recommends default values of the dissolution parameters for three generic reference

absorption types: fast (F), moderate (M) and slow (S) (ICRP, 1994). However, it has long been

recognised that the default assignment of uranium compounds to these reference types often did

not reflect other specific information on their actual absorption characteristics and could lead to the

production of implausible estimates of intake and dose (Riddell, 1995). In the revised HRTM (ICRP,

2015a; ICRP, 2015b), new values are proposed for uranium forms assigned to the reference

absorption type. These new material specific parameter values derived from a review of uranium

pulmonary absorption data offer greater fidelity to the actual absorption characteristics of these

materials, resulting in much better estimates of intake and dose. In the absence of more specific

information, one of the reference types can be chosen according to the possible chemical form of the

radionuclide.

There is often little or no information available on the absorption parameter values from

measurements made of materials at the specific facilities of interest over the time periods

considered by this study. Cohort-specific information on lung parameters is provided in the annex of

the CURE deliverable D2.2. on site-specific procedures (Blanchardon et al., 2014a). If the uranium

compound to which a worker is exposed is known then the absorption parameter values of table 6 of

CURE deliverable D2.2. will be applied (Blanchardon et al., 2014a). When less precise information is

available, one of the reference F, M or S absorption Types or a mixture of them can be assumed. If no

information is available, an equal mixture of the three reference Types will be used. If the worker is

46

possibly exposed to both Types F and M, both Types M and S, or both Types F and S compounds, a

fifty-fifty mixture of the two possible Types will be assumed. Table 3 indicates parameter values that

provide a reasonable approximation of the expected biokinetics of a mixture of reference Types and

will be used in intake and dose calculation. The default parameter values may be overridden when

more specific information is available on the absorption of uranium compounds at a given facility or

workplace.

Table 3: Absorption parameter values representing mixtures of ICRP reference Types (arithmetic

mean of default parameter values for absorption Types from (ICRP, 2015a)).

Mixture fr sr (d-1) ss (d

-1) fA

F+M+S 0.403 5.33 2.55 x 10-3 8.07 x 10-3

F+M 0.60 6.50 5.0 x 10-3 0.0120

M+S 0.105 3.0 2.55 x 10-3 2.10 x 10-3

F+S 0.505 6.5 1.0 x 10-4 0.0101

Reconstruction of workers histories

The doses will be calculated for workers monitored for uranium exposure through urine analyses on

or after a defined date which will be chosen as the earliest date for which reliable bioassay data are

available.

Workers employed at more than one participating facility or employed more than once at the same

facility will be identified and their occupational and dosimetric history will be reconstructed. External

and internal radiation doses received before employment in a participating facility, and in non-

participating facilities, will be obtained where available.

Some of the information required for dose reconstruction relates to the characteristics of the

plant/workplace/activity and may be entered only once and applied to all workers involved in the

workplace/activity. This includes the characteristics (chemical form, solubility type, etc.) of the

isotopes present in various work environments and the measurement techniques implemented

within specific time periods. On the other hand, some data will be specific to a worker. Where

feasible the exposure and its parameters will be defined by combining administrative files (successive

jobs in the career) with a job-exposure matrix (JEM) and a registry of incidents. Workplace specific

aerosol parameters may also be determined from a representative group of workers with sufficient

bioassay measurement data. With regard to the type and time of intake, a constant chronic intake

regime is assigned to each period of routine exposure during employment and acute intakes are

defined according to records of incidents and special monitoring (e.g. in-vivo, faecal results).

4.3.4. Monitoring data

4.3.4.1. Uranium miners and millers

Uranium and other long-lived radionuclides

Exposure to long-lived radionuclides (LLR) has been measured in mines in terms of gross alpha activity (h Bq m-3). The radionuclides considered include:

47

238U, 234U, 230Th, 226Ra, 210Pb and 210Po of the 238U chain

235U, 231Pa and 227Ac of the 235U chain

232Th, 228Ra, 228Th and 224Ra of the 232Th chain.

The assumed activity ratio of 235U/238U is 5/95 and the assumed activity ratios of 232Th/238U in different mines are given in (Marsh et al., 2009). It is also assumed that 25% of the 222Rn gas and 220Rn escapes from the ore dust particles (details for radon and its progeny are provided below). Because of its short radioactive half-life (4 s), 219Rn is assumed not to escape from the ore matrix. Under these assumptions about 7.6 alpha disintegrations occur per becquerel of 238U. Thus, the incorporated activity of 238U (Bq) is given by the gross alpha activity exposure (h Bq m-3) multiplied by the breathing rate (m3 h-1) and divided by 7.6 (Marsh et al., 2008).

For millers processing non-ore material, it is assumed that only the uranium nuclides were present in

the material with activity ratios typical for natural uranium (i.e. activity ratio of 235U/238U is 5/95 and

that of 235U/238U is 1). Therefore, to interpret the gross alpha activity measurements, it is assumed

that 2 alphas are emitted per disintegration of 238U.

Radon and progeny

The historical unit of exposure to radon progeny applied to the uranium mining environment and

available in the Czech, French and German cohort databases is the working level month (WLM),

defined as the cumulative exposure from breathing an atmosphere at a concentration of 1 working

level (WL) for a working month of 170 hours. A concentration of 1 WL is any combination of the

short-lived radon progeny in one liter of air that will result in the emission of 1.3 105 MeV of

potential alpha energy. To calculate the absorbed doses per WLM, the activity concentration (in h Bq

m-3) of each of the short-lived radon progeny (218Po, 214Pb, 214Bi) corresponding to 1 WL is required.

These concentrations for 1 WL can be uniquely determined from the equilibrium factor, F which is a

measure of the degree of disequilibrium between the radon gas and its progeny (ICRP, 1993). A value

of 0.4 for F is typical for a mine and the activity ratios assumed are consistent with the assumptions

of the NRC (NRC, 1991).

External gamma

The dose from external gamma exposure is readily available in the databases and recorded in mGy.

The operational quantities measured by dosimeters are converted into organ absorbed dose by the

application of correction factors taking into account the attenuation of the external radiation

spectrum by body tissues, as indicated in 3.2.

4.3.4.2. Uranium workers

Bioassay measurement results and normalisation

The data to be used in the dose assessment are values of uranium activity excreted per day (Bq/d).

Bioassay measurement results recorded as Bq/day or µg/day or otherwise indicated as already

representing daily excretion will be kept as they are. Other bioassay results will be converted to a

mass or activity of uranium excreted per day through normalisation by volume (using reference daily

excretion values of 1.6 L for males and 1.2 L for females) or by creatinine content (using reference

daily excretion values of 1.7 g for males and 1.0 g for females) depending on the information

available on the sample. Mass values for uranium will be converted to activity using any available

information or reasonable assumptions about the isotopic composition of the exposure material.

48

Detection limit and reporting limit

The sensitivity of a monitoring technique is defined by its detection limit (DL). Uranium analytical

techniques are relatively sensitive and, for some, the limit of detection could potentially be less than

excretion from normal dietary intake of uranium in some areas. In order to discriminate between

non-occupational, i.e. dietary, excretion of uranium and that resulting from occupational exposures,

a ‘reporting limit’ (RL) is sometimes employed. Results less than the detection limit or the reporting

limit are usually recorded only as being less than this limit, without indication of the raw

measurement value. However, more recent measurements of excretion in non-exposed persons

living in the same location as those potentially occupationally exposed to uranium at various sites

has indicated that normal dietary excretion levels can be much lower than the relevant RL. Results

from these surveys, at various sites, of actual levels of uranium in urine arising from dietary intake,

can be used to supplement/correct occupational monitoring data.

Scattering factors

The overall uncertainty of measurement may be described by a log-normal probability distribution

which geometric standard deviation is called a scattering factor (SF) (Marsh et al., 2007). For in vitro

measurement, the value of SF depends on the sampling procedure. Several of the participating

institutions have calculated SF for their own measurements, including values related to early

measurement techniques. Where such data are absent, the values elaborated by Working Group 7 of

EURADOS and published in the IDEAS guidelines (Castellani et al., 2013) are to be used.

External gamma

The dose from external gamma exposure is readily available in the databases and recorded in mGy.

The operational quantities measured by dosimeters are converted into organ absorbed dose by the

application of a correction factor taking into account the attenuation of the external radiation

spectrum by the body tissues, as indicated in 3.2.

4.3.5. Assessment procedure

4.3.5.1. Miners and millers

For miners and millers, the intake is calculated as the product of an activity concentration of a

radionuclide in the air by the breathing rate (considered in the “reference miner type” as explained

above) and by the time spent breathing this contaminated air (time integrated concentration). The

dose, calculated using the aforementioned biokinetic and dosimetric models, is proportional to the

intake and depends on the physico-chemical parameters of the radionuclide. For uranium and long-

lived radionuclides, a constant chronic inhalation for the year of exposure is assumed. Because of the

long half-lives and the long retention within the body, the inhaled LLR in the ore dust continue to

give a dose to the organs following exposure. This needs to be accounted for when calculating the

annual doses to a miner over a long period of time. For radon and progeny, an acute inhalation

intake regime is considered because of the short half-lives.

4.3.5.2. Uranium workers

Maximum likelihood approach

49

The intake of radionuclides for an individual worker will be evaluated using the identified likely

pattern of exposure (i.e. “intake regimes” - a set of chronic exposure periods and/or acute exposure

incidents), the relevant biokinetic model and parameter values for each intake regime, and the

available bioassay data for the individual. A maximum likelihood approach will be used to assess

intakes. It derives intake value(s) that maximise the probability of observing the bioassay

measurements, both real and censored values, under the hypothesis of the measurement error

model specified.

Setting intake regimes

The assumed intake regime will consist of one or more periods of constant chronic intake, depending

on the work history, plus any acute intakes to be identified from operational data. Not all reported

incidents lead to acute intakes. Any incident that resulted in follow-up bioassay monitoring (i.e.

special sampling or in-vivo measurement) being performed will be assigned an acute intake date if

that monitoring yielded positive results.

The chronic intakes will then be assigned to each phase of a worker’s career involving a potential risk

of internal exposure. Just one chronic intake is required if the worker did the same job in the same

facility throughout his career, but a sequence of chronic intakes is required for a more complex work-

history. A job-exposure matrix is one possible way to specify the periods of constant potential

exposure within a work history. Bioassay monitoring is considered as the most reliable indication of

potential exposure. The assumed period of chronic exposure would therefore go from one

monitoring interval before the first measurement data (or from the date of first employment if this is

later) until the day of the last measurement data (or the end time of professional activity). A break in

bioassay monitoring over more than 3 monitoring intervals would indicate the cessation of potential

exposure during that break and no intake would be evaluated during that period. More precise

information is to be used when it is available.

Specification of biokinetic and error models

The biokinetic models indicated above will be applied. By default, intake is assumed to take place by

inhalation and is represented by the human respiratory tract model (ICRP, 2015a). If there is

indication of a wound intake, the wound model (NCRP, 2006) is to be applied. The particles cleared

from the airways to the gut and the unlikely ingestion intakes will be considered within the human

alimentary tract model (ICRP, 2006).

The particle size, pulmonary absorption type and wound retention category are fixed as explained

above. Plant-specific or incident-specific model parameter values will be used when available.

Aforementioned generic default values will be used in other cases (Blanchardon et al., 2014a). In

some cases it may be possible to infer individual-specific model parameters, where sufficient data

(e.g. faecal, in-vivo, air-sampling results) are available.

The measurement error model is log-normal with a geometric standard deviation, or scattering

factor (SF), depending on the monitoring technique as explained above. For results at or below the

limit of detection of a technique, then errors due to counting statistics may predominate and a

normal distribution may be used to describe errors.

Periods of chronic exposure with only censored bioassay data

50

When all bioassay data within a chronic exposure period are less than the detection limit (DL) or

reporting level (RL), two intake values will be calculated for that period: (1) a minimum value of zero

and (2) a maximum value calculated by setting the last bioassay data as positive and equal to the

value of the DL/RL, while other bioassay data remaining censored. As a consequence, the doses for

the following years will be calculated as a range from a minimum dose to a maximum dose,

corresponding to the minimum and maximum values for former intakes.

Workers with bioassay data but no positive (above threshold) results could represent an important

part of the study population and their exclusion from the epidemiological study might lead to

selection bias (although this is to be evaluated in each cohort, by comparing the characteristics of

these workers with those having at least one positive result) and in this case, affect resulting risk

estimates. For those workers, annual doses will thus be calculated as a range from 0 to a maximum

value.

4.3.5.3. Quality assurance

The dose assessments for each cohort will be made by the dosimetrist associated with that cohort.

In order to ensure consistency in approach between the dosimetrists and to cross-check the results

of the various dose assessment software packages, samples of the assessments will be exchanged

between the dosimetrists.

51

4.4. Methods for statistical analyses

Three forms of analyses are envisaged:

The first (“broad-base”) analysis will generate Standardised Mortality Ratios (SMRs) for a

range of diseases (both cancer and non-cancer) for which sufficient cases are observed

and/or that are of a priori interest regarding the suspected effects of uranium.

The aim of the second analysis will be to estimate a dose-response relationship between

uranium dose and disease risk.

The aim of the third analysis will be a preliminary exploration of the associations between

uranium exposure and the biomarkers selected for the study

Based on current knowledge about the biokinetics of soluble and insoluble uranium types,

information on biological effects stemming from animal experiments and suggested effects from the

few available epidemiological studies (see sections 1.3. and 1.4. for details) the diseases of interest to

the first two types of “classical” epidemiological analyses will be as follow:

Lung cancer

Lymphatic and haematopoietic cancers

Kidney cancer

Bone cancer

Digestive tract cancers

Cardiovascular diseases

Neurological diseases

Non-malignant respiratory diseases

Non-malignant renal diseases

These outcomes will be studied using mortality data. However, incidence data will also be available

for cancer outcomes in some cohorts. In addition, broad groupings of diseases or causes of death will

be examined:

All causes

All solid cancer

All solid cancer excluding lung cancer (with regard to miner studies)

Cancers related to smoking

Cancers not related to smoking.

Biomarkers of interest for analyses have been presented in section 4.2.1.

4.4.1. Broad base analyses

Two main pooled datasets will be available: one for miners/millers, and one for nuclear workers

(others may be defined for sensitivity analyses).

Standardised Mortality Ratios (SMR) will be computed in order to compare the mortality rates (from

the causes of death listed above) estimated from these pooled cohorts to the ones estimated in the

general populations from which the cohort members are drawn. As these analyses do not require

52

individual dose measurements for each subject, they will allow including the largest number of

subjects from each cohort. This will maximise the statistical power of the analyses to identify which

diseases are in excess in the cohorts.

Among nuclear workers, separate SMR values will be computed for those who were only exposed

externally to radiation and for those who were additionally (or instead) exposed internally to

uranium, and the ratio of these SMR’s will be computed. Any statistically significant difference

(deviation) of this ratio from one will indicate that uranium exposed workers are at different risk to

those only exposed to external sources of radiation. For miners and millers who were all exposed to

both external gamma radiation and uranium, the comparison will be made with the general

population only.

A range of additional sensitivity analyses may also be performed, for example:

Analysis of variation of SMRs with exposure to other nuclides e.g. plutonium in addition to

uranium

Analysis of variation of SMRs with age at first exposure, time since exposure, duration of

employment, between cohorts and countries.

Analysis of the impact of confounding factors. Examination of confounders would be

dependent on the information available in the cohorts with sufficient coverage. Possibilities

include stage of fuel reprocessing (or levels of enrichment), socio-economic status and in

some cohort subsets, contact with hazardous chemical or smoking. Even if smoking data are

not available for the whole cohorts, the risks for diseases potentially related to smoking

(IARC, 2004) can be compared to the risks for non-smoking related diseases in all cohorts.

The main focus of the pooled analyses will be on mortality, since mortality data (dates and causes of

deaths) will be available for all cohorts. However, it will also be possible to conduct similar analyses

with cancer incidence data (by computing Standardised Incidence Ratios) in countries where cancer

registries are available (United Kingdom and Belgium).

4.4.2. Dose-response analyses

The aim of these analyses will be to study the relationships between absorbed dose from uranium

and mortality from specific diseases or – where available –cancer incidence, accounting for potential

confounders including exposure to external gamma radiation exposure and other internal nuclides

than uranium. It will also aim to estimate separately external and uranium dose-response

relationships and to compare them. In doing so, it will be possible to empirically estimate the

radiation quality effect for comparison with the standard value for internal alpha radiation exposure

as proposed by (ICRP, 2007).

These analyses will initially be performed separately for the nuclear workers and miners/millers

groups. A final stage will be to compare the results between the nuclear workers and the

miners/millers to determine whether a single consistent estimate of uranium risk (and a single

radiation quality estimate) can be defined using a single pooled cohort of all the compatible workers

and miners/millers cohorts.

53

Ideally, Cox regression will be fitted to datasets providing information at individual level. However, in

the event that permission is not given to share data between partners at the individual level in a

timely fashion, standard Poisson regression will be rather used to fit dose-response relationships

from aggregated data (i.e., at a stratum level pooling homogeneous person-years according to sex,

attained age, calendar period, and other factors).

Where feasible, it would be also useful to perform further statistical analyses to study the potential

modifying impact of both time-dependent factors (e.g., age at exposure, time since exposure…) and

potential confounders (e.g., smoking, occupational exposures other than uranium) on the dose-

response relationships of interest. These analyses will be performed on groups of frequently

occurring diseases, e.g. all solid cancer, leukaemia excluding or including chronic lymphocytic

leukaemia, smoking and non-smoking related cancers, and on such individual cancer and non-cancer

disease groups in which a significant number of occurrences are observed (these are likely to include

lung cancer and circulatory system diseases).

4.4.3. Exploratory analysis of biological information

Because the primary objective of pilot studies for molecular epidemiology will be to determine

whether the standardized operating procedures (SOPs) proposed as part of the CURE project can be

implemented in an effective way in a prospective cohort, in the first place biological data will only be

available for a cross-sectional sample of workers.

Exploratory statistical analyses will focus on biomarkers measured in two subgroups of this cross-

sectional sample. One subgroup will be selected to have no (or low) exposure to uranium while the

other will be selected to be similar in other respects but to have high exposure to uranium. Standard

multivariate statistical analyses will be performed to determine potential differences in biomarker

measurements between the two groups.

Specific methods such as Partial Least Squares Regression-Discriminant Analysis (PLS-DA) may be

used for to the analysis of multidimensional data (e.g.: analyses of metabolomics data taking

potential confounders into account)(Bonvallot et al., 2013).

54

4.5. Characterization and propagation of uncertainties

4.5.1. Introduction

Accounting for uncertainties in dosimetry and in dose-response models used in epidemiologic studies

of uranium workers and miners/millers is a complex issue and a vast field of investigation. This is due

to the complexity of dose assessment procedures, but also to the various sources of uncertainty that

can be identified (even if, some are of arguably less importance than others, see the UWG final

report (Giussani et al., 2014)) and to the advanced statistical methods that are needed to deal with

such uncertainties.

Research has been conducted, notably by partners of the CURE consortium, on the uncertainties

related to the quantification of radiation doses due to internal exposure to radionuclides (Davesne et

al., 2010; Li et al., 2011; Puncher et al., 2013). The impact of specific sources of uncertainty such as

exposure measurement error on the disease risk estimates is a relatively new but developing field in

radiation epidemiology (UNSCEAR, 2012), and several partners of the CURE project have begun to

explore it, e.g.: (Allodji et al., 2012; Hoffmann et al., 2014).

Several sources of uncertainty as well as their respective nature and possible impacts on dose and

risk estimates have been identified at several steps of the proposed scientific project (e.g.:

interpretation of bioassay, reconstruction of exposure conditions, dose calculation process, health

outcome ascertainment, epidemiological and biomarker analyses), as detailed in the UWG final

report.

Based on the uncertainty matrix developed by the UWG, but also of case studies conducted by UWG

members and of their experience of as part of various projects (see the UWG final report), it has

been possible to prioritize research needs in the field of health risk assessment pertaining to

occupational exposures to uranium. The following propositions should be considered as a partial

road map for a middle-to-long-term strategic work plan. It is not intended that all these research

questions are addressed within the scope of a single research project.

The priority issues to address were identified as follow:

Uncertainties on the lung solubility of uranium compounds encountered in some

occupational settings. Of all the parameters employed in the biokinetic models necessary for

retrospective reconstruction of internal radiation doses, those governing absorption of

radionuclides from the lungs to blood have the greatest impact on estimates of lung dose. It

is acknowledged that the impact on doses to other biokinetic and dosimetric endpoints than

lung will be more limited.

Censored bioassay data (i.e. measurement below DL or RL).

The identification of suitable statistical methodologies to account for the uncertainty

inherent in estimates of dose when estimating radiation-induced disease risk.

In order to address the aforementioned issues, the following research perspectives have been

proposed:

Characterizing the best estimates and probability distributions of model parameters and

other relevant uncertainty sources

55

Testing the feasibility to account for the uncertainty inherent in estimates of dose when

estimating radiation-induced disease risk.

Creating a synthetic cohort as a tool for testing the proposed methodologies

These research perspectives will be developed below:

4.5.2. Characterizing the best estimates and probability distributions of model parameters

and other relevant uncertainty sources

Deriving best estimates of parameter values is required whether the primary dose deliverable is

point estimates of dose or doses in the form of Bayesian (or other) probability distributions obtained

from uncertainty analyses. For these reasons significant effort should be directed into deriving prior

distributions for key parameters, particularly those representing “reducible” uncertainties (identified

in the uncertainty matrix (see the UWG final report)). Again, of all the biokinetic parameters, it is

those governing absorption from lungs to blood that have the greatest impact on estimates of lung

dose.

Bayesian methods to estimate uncertainties on doses were developed at PHE as part of the EU

funded projects Alpha-Risk and SOLO and under the US-funded JCCRER projects for Russian Mayak

workers. One aspect of this approach consisted in deriving informative prior probability distributions

for exposure parameters from data that directly relate to the workers of interest and the material

they were exposed to. In the dose estimation for Mayak workers, Bayesian inference was applied to

obtain best estimates of the dissolution parameters for plutonium nitrate and oxide compounds

from worker bioassay and autopsy data. The output from these calculations consisted in probability

distributions of absorption parameter values that were then applied to the whole cohort to calculate

uncertainties on doses. In the same way, other sources of uncertainty, such as aerosol size and

dispersion parameters, and likely intake levels, could be obtained from a review of workplace

monitoring data.

Similar approaches could be used to derive estimates of the absorption parameters for uranium

materials. Distributions (representing variability) derived for particle transport clearance rates and

other physiological parameters for the Mayak workers could be used as a reasonable starting point

for deriving distributions for individual specific parameters for the cohorts considered in CURE. It can

indeed be assumed that these distributions are likely to be material independent and similar across

cohorts. However, scope exists to further refine these distributions using relevant uranium worker

autopsy data, if available.

Therefore, priority should be given to obtaining best estimates of the rate of absorption of the

specific uranium materials that workers were exposed to and likely default intake rates; hence,

appropriate (informative) bioassay data from workers within the cohort will be identified for

Bayesian analysis to determine absorption rates. Failing this, the goal should be deriving reasonable

default values for exposures where the type of material is unknown.

Further information might come from a pilot study on biomarkers in sputum, saliva or nasal swabs. A

possible outcome of such work could be the identification of biological markers useful for the

characterization of lung solubility parameters and consequently for reducing the uncertainty on the

assumptions used in exposure/dose assessment.

56

Other sources of uncertainty such as qualitative modelling hypothesis (e.g.: modelling of activity

measurement error, correction for contribution of dietary intakes of uranium to bioassay) should

also be characterized in order to be integrated in a full uncertainty study.

4.5.3. Testing the feasibility to account for the uncertainty inherent in estimates of dose

when estimating radiation-induced disease risk

The development of an innovative approach to derive uncertainty on risk estimates from

uncertainties arising from the different sources identified in the matrix (see the UWG final report) is

a major challenge. The question of integrating dosimetry and epidemiology can be considered as one

global issue or, alternatively, as a succession of two smaller issues: 1) estimation of dose and its

associated uncertainty from uncertain exposure data; 2) evaluation of the risk and its uncertainty

from dose estimates and their uncertainties.

Several methods to account for the uncertainty inherent in dose estimates when estimating disease

risk have been discussed and proposed in the UWG final report.

All the proposed approaches need to be further evaluated, and their feasibility and validity in an

epidemiological study are still to be verified. Therefore, their application to the CURE cohorts would

not be realistic in a short time frame. Nonetheless, feasibility studies could be conducted using

smaller sets of representative subjects and then extended to all workers or miners/millers having

similar individual characteristics or experiencing similar conditions of exposure and monitoring.

Alternatively, a purposely created synthetic cohort (see below) could be used. These studies would

give important indications on the possible drawbacks and difficulties to be encountered in the

implementation of the methods to the full CURE cohorts.

4.5.3.1. Handling probability distributions representing uncertainties on doses as input data

for propagation in epidemiological analyses

A previous experience as part of the Alpha-risk project has showed that propagating distributions

reflecting both shared and unshared uncertainties on dose estimates (derived by a theoretically

correct Bayesian retrospective dosimetric framework) throughout epidemiological analyses can lead

to a series of difficulties, notably numerical problems due to large inter-/intra-cohorts variations and

scaling problems when manipulating the data due to different levels of apparent informativeness

between cohorts (and also within one cohort). However, it appears that if the priors are optimised

with respect to the bioassay data and, if the bioassay data then provide no additional information

regarding model parameter values (beyond that already specified by the priors), then the method

can be simplified in such a way that convergence can be achieved while preserving the shared error

structure. Nevertheless, this methodology is still in its infancy and needs to be further evaluated.

4.5.3.2. Determination and use of simple summary statistics to represent uncertainty on

dose estimates

A possible alternative approach could be to account for uncertainty on dose estimates through simple summary statistics: confidence or credible interval and standard deviation for example. Even though they are less informative, these summary statistics could be more easily used than a probability distribution on dose to account for dose uncertainty when estimating disease risk from dose-response models.

57

4.5.3.3. Directly accounting for exposure uncertainty when estimating risk

A Bayesian approach to account for exposure measurement error when estimating the risk of cancer

is currently under development at IRSN. This methodology derives risk directly from exposure and its

uncertainty. The main idea is to combine conditionally independent sub-models into a unique

framework, called Bayesian network, and then to infer the whole model under the Bayesian

paradigm, using, for instance, a Monte-Carlo Markov Chain algorithm. The independent sub-models

are:

a disease model describing, for each worker, the relationship between the true (latent)

radiation exposure and the time until death caused by that specific disease

a measurement model describing, for each worker, the conditional probability distribution of

his true (resp. observed) exposures given his observed (resp. true) exposures in case of

Berkson (resp. classical) measurement error respectively

an exposure model describing, in case of classical measurement error only, the probability

distribution of the true exposures.

This so-called Bayesian structural approach is based on a unique and coherent framework in which all

the parameters of the measurement, exposure and disease models are estimated simultaneously

allowing radiation exposure uncertainty to be propagated into the risk parameters estimation. This

approach is currently being applied to refine the risk estimates for lung cancer due to radon

exposure in a uranium miners’ cohort. It could be extended and specifically adapted to the case of

uranium exposures of miners, millers and other nuclear workers.

4.5.4. Creating a synthetic cohort as a tool for testing the proposed methodologies

A so-called "synthetic cohort" can be created by generating simulated dosimetric and health

outcome data for a number of artificial subjects. Such a synthetic cohort would be used for a number

of purposes. A primary use would be evaluation and validation of methods currently used to

investigate uncertainties in dosimetric data. Since the cohort could be constructed to have a

particular risk and dose-response relationship, this would also allow a sensitivity analysis to be

carried out to quantify impacts of dosimetric uncertainties on measures of risk estimated by

epidemiologists. One significant advantage of using a synthetic cohort is the potential for sharing all

data (dosimetric and epidemiological databases) between members of a future project without the

usual constraints of data protection and ethics. A number of issues might be explored:

The possibility of using individual dose distributions for each member of the cohort within

analyses

Impact on risk estimates of attempting to account for both shared and unshared

uncertainties

Potential for combined analyses of cohorts with dosimetry of different levels of

informativeness

How to account for the reliability of assumptions used in the development of a Job Exposure

Matrix

58

5. CONCLUSIONS

The CURE project has reached its objective to propose an integrated research proposal, thanks to an

effective collaboration and strong interactions between epidemiologists, biologists, dosimetrists and

biostatisticians. The CURE consortium proved to be a highly productive think-tank for future projects.

The work conducted has several major strengths:

The multidisciplinary approach adopted as part of CURE, integrating dosimetry,

epidemiology, biology/toxicology and biostatistics proved to be scientifically pertinent. It

allowed producing a research protocol with strong potential to improve both the

understanding and the quantification of the biological and health effects of uranium

exposure.

CURE has built a strong multidisciplinary consortium of epidemiologists, dosimetrists,

biologists, toxicologist and biostatisticians, conducting top-level research in their respective

fields. These researchers brought together a broad array of complementary skills, developed

understanding of their respective, and mutual, contributions. This aspect was essential to the

collective construction of the research project. This has prepared the consortium to the

implementation of future multidisciplinary research projects on the biological and health

effects of internal contaminations.

Thanks to the extensive experience of CURE partners and of invited experts, the

opportunities for future projects, but also their potential pitfalls, have been identified. This

allowed for the definition of an ambitious but realistic protocol, which can be implemented

in the short term.

The preparation work conducted as part of CURE will make it possible to conduct the first

pooled epidemiological analysis of uranium workers and miners in Europe. This will include

large cohorts with long follow-up (providing optimal statistical power to detect increased

risks), in which internal and external dose estimates are either readily available or are about

to be. Such a pooled epidemiological analysis will have potential to deliver results directly

relevant to radiation protection.

A full protocol has been prepared that will allow to test the integration of biological

indicators into epidemiological studies of uranium exposed workers, by conducting pilot

molecular epidemiology studies. This is a crucial step on the road map to the integration of

modern radiobiological and epidemiological research. The pilot studies will be nested within

established cohorts, which will allow capitalizing on carefully collected information

(including, but not limited to, dosimetric data). This will ensure both a high data quality and

cost effectiveness.

Contacts have been established with research teams involved in similar research projects

outside Europe (USA, Kazakhstan, Russia), as well as with experts from various fields of

expertise within Europe, notably in molecular epidemiology and biobanking. These

59

contributions proved to be very effective and the establishment of contacts with external

experts will be pursued in future projects.

Early dissemination work has been conducted, throughout oral and poster communications

in conferences and workshops (see annex 2). Further dissemination will be ensured by

submitting scientific papers directly resulting from the CURE project to peer reviewed

journals.

As stated above, a number of potential pitfalls have been identified by the CURE consortium, and

these will be anticipated.

Obtaining agreements from ethics committees (ethics of biological sampling, storage and use

of samples, data access, data protection) can be time consuming. Procedure vary from

country to country, but in each of them delays have to be anticipated. For all CURE cohorts,

the necessary agreements from ethics committees have already been collected, or

applications have been submitted and are currently being reviewed. This anticipation will

allow for the implementation of the proposed research protocol to be started in the short

term (i.e., before year 2016).

Obtaining agreements from stakeholders for molecular epidemiology studies (employers,

doctors, workers, worker representatives committees) can be challenging in some settings.

Biological sampling is only possible if there is interest and support to the project by the

workers who will provide biological samples, and by the medical centres where samples have

to be collected. Agreements from employers are needed if sample collections are organized

during medical check-ups. The local acceptability may depend upon the social and economic

context. The personal views of occupational physicians about the study goals will also

determine their willingness to authorize certain kinds of biological sampling (e.g., sputum

collection). Communication with all stakeholders (workers, worker representatives, doctors,

employers) is therefore a key issue. An information sheet was developed as part of the CURE

project to help in this process. For the pilot study proposed in Rozna (Czech Republic), the

agreements from all stakeholders except workers (at this stage), have already been obtained.

The individual consent to participate from voluntary workers will need to be obtained as part

of pilot studies.

The feasibility to set up molecular epidemiology studies in some settings also depends on

other practical constraints. The proposed biological sampling scheme needs to be compatible

with the usual organization of medical check-ups, and with local infrastructures. Last, in

some settings only a limited quantity of blood can be collected for the purpose of a study,

depending on the volume already collected for the purpose of routine surveillance. All these

conditions were carefully checked as part of the preliminary feasibility studies conducted in

the study settings proposed for pilot studies, and the logistic strategies were defined

accordingly.

The use of biomarkers in epidemiological analyses is a developing field. It is acknowledged

that the proposed list of biomarkers might not cover all the potential biological effects of

uranium. However, the list of proposed biomarkers can evolve in the future. It is open for

60

emerging biomarkers of interest, since the collection and proper storage of biospecimen

(biobanking) will give the opportunity for a multitude of future analyses.

It is acknowledged that the levels of exposure to uranium of workers currently active in

Western Europe are not representative of those encountered in the same settings a few

decades ago. It might be relevant in the future to launch other pilot studies in other settings

located outside Europe, where workers have been exposed to uranium at higher levels

during recent years.

Sources of uncertainties have been identified at several steps of the proposed project.

However, the work conducted by the UWG as part of CURE allowed identifying the sources

of uncertainty which should be addressed in priority, as opposed to others that are

considered to be of lower importance. Further work will be conducted to better characterize,

and whenever possible, reduce uncertainties. Methodologies have been proposed to

account for the uncertainty in estimates of dose when estimating radiation-induced disease

risk, and these will be evaluated as part of future projects.

Funding is to be found to apply the research protocol designed during the CURE project.

Financial support from the European Commission will be a major condition for launching

future projects. Nevertheless, the Commission will not be considered as the only source of

potential funding. Screening of potential complementary sources of funding at national and

international level will be regularly evaluated by each partner.

In the short term (i.e., within the next two years), major steps of the protocol developed as part of

CURE can be implemented. Depending upon availability of funding, it would be possible:

To complete or wherever needed, to improve internal dosimetry within established cohorts

of uranium workers and miners in Europe.

To estimate health risks associated with internal doses resulting from uranium exposure in

the pooled cohorts, which would be directly informative for radiation protection.

To better characterize the sources of uncertainties identified as priorities during the CURE

project, to reduce these uncertainties wherever feasible, and to test methodologies to

account for the uncertainty in estimates of dose when estimating radiation-induced disease

risk.

To set up pilot molecular epidemiology studies. This would allow : 1) testing the collection of

biological samples and of related information, which will provide precious feedback for

future projects. 2) testing the associations of several biomarkers with internal and external

radiation dose. Targeted approaches would provide information on the mechanisms of

biological responses to radiation at low dose. Non targeted approaches such as

metabolomics would be tested for the first time in humans, in relation with chronic exposure

to internal emitters. This would allow assessing whether new pools of biomarkers could be

identified as biological signatures of exposure in humans. Such signatures were recently

61

observed in rats exposed to uranium (Grison et al., 2013) and cesium (Grison et al., 2012). 3)

initiating the creation a biobank that would be an invaluable resource for future integrated

studies on the effects of chronic exposure to internal emitters and more generally, a

strategic investment for radiation protection research.

To further develop contacts established with other research teams interested in the

biological and health effects of uranium exposure outside Europe (USA, Kazakhstan, Russia,

Canada). This would include assessing the feasibility of pooling established epidemiological

cohorts in these countries with CURE cohorts or to set up new ones, but also of setting up

pilot molecular epidemiology studies in these countries.

To create closer links with research teams and organizations outside the field of uranium

research. This would include research on the effects of other internal emitters, as well as on

other exposure situations than occupational exposure. Exchanges with the MELODI,

EURADOS, ALLIANCE and NERIS platforms would be organized to better identify, and

wherever feasible to build on, possible synergies. Exchanges with researchers outside of the

radiation field would be organized, notably on the topic of molecular epidemiology, systems

biology, and advanced statistical approaches for the propagation of uncertainties in

regression models.

In order to complete the steps listed above, the CURE consortium will submit a proposal for funding

to the European Commission. The impacts of the proposed work would extend far beyond the issue

of uranium exposure in workers and miners, since the above steps are crucial for:

The improvement of knowledge on the health effects of exposure to internal emitters.

Especially, producing information about the magnitude of risks related with internal alpha,

compared with external gamma, radiation will help assess the validity of the current value

for the radiation weighting factor for alpha emitters (wR)(ICRP, 2007).

The integration of modern radiobiological and epidemiological research, which is a major

goal for future radiation protection research. A proof of principle is needed for this approach.

The successful constitution of a modern biobank and/or the validation of biomarkers of

exposure or effects would create important precedents. They would open the way to the

creation of large-scale international biobanks. In the future, such biobanks would become

major infrastructures for integrated radiobiological and molecular epidemiology research on

the effects of chronic exposure to internal emitters and more generally, for radiation

protection research. The proposed project would allow defining the bases of a road map

toward the integration of modern radiobiological and epidemiological research.

This proposal will build on at least 3 DoReMi tasks: CURE (task 5.8.), IntEmitUM (task 5.5.1) and

AIRDoseUK (task 5.5.2.). It will directly address research priorities identified by the HLEG (dose-

response relationship at low dose/low dose rate exposures for cancer; non cancer health effects;

internal exposure; influence of radiation quality). It will also addressing research priorities mentioned

in the Strategic Research Agendas of MELODI (again, dose and dose rate dependence of cancer risk;

improvement of the understanding of the mechanisms contributing to radiation risk at low dose and

dose-rate exposure; non-cancer effects; integration of biological approaches for radiation risk

62

evaluation in epidemiological research; effects of and risks associated with internal exposures;

differing radiation qualities and inhomogeneous exposures) and EURADOS (improvement in the

modelling of biokinetics and dosimetry of internal emitters, improvement of uncertainty evaluation

of doses, dosimetry for historical cohorts).

In fine, future projects building on reflections conducted as part of the CURE project will be able to

improve the estimates of risk related to exposure to uranium, but also of other incorporated

radionuclides and more generally of low doses of ionizing radiation, throughout to the integration of

dosimetry, biology, epidemiology and biostatistics. In the future, it might be envisaged to open the

reflections to other disciplines interested in the effects of internal contaminations by radionuclides.

63

REFERENCES

Abilmazhinova A, et al. Conference Paper: Gene expression profiles in uranium industry workers of Stepnogorsk city occupationally exposed to ionizing radiation. European Human Genetics Conference. Milan, Italy May 2014. (2014).

Allodji RS, et al. The performance of functional methods for correcting non-Gaussian measurement error within Poisson regression: corrected excess risk of lung cancer mortality in relation to radon exposure among French uranium miners. Statistics in medicine (2012) 31:4428-4443.

Ansoborlo E, et al. Determination of the physical and chemical properties, biokinetics, and dose coefficients of uranium compounds handled during nuclear fuel fabrication in France. Health physics (2002) 82:279-289.

Ansoborlo E, Lebaron-Jacobs L, Prat O. Uranium in drinking-water: A unique case of guideline value increases and discrepancies between chemical and radiochemical guidelines. Environment international (2015) 77C:1-4.

Araujo JA, Nel AE. Particulate matter and atherosclerosis: role of particle size, composition and oxidative stress. Particle and fibre toxicology (2009) 6:24.

Atkinson WD, Law DV, Bromley KJ. A decline in mortality from prostate cancer in the UK Atomic Energy Authority workforce. Journal of radiological protection : official journal of the Society for Radiological Protection (2007) 27:437-445.

ATSDR. Toxicological profile for uranium (2013) Atlanta, Georgia (USA): Agency for Toxic Substances and Disease Registry.

Bal W, Protas AM, Kasprzak KS. Genotoxicity of metal ions: chemical insights. Metal ions in life sciences (2011) 8:319-373.

Bečková V, Malátová I. Dissolution behaviour of 238U, 234U and 230Th deposited on filters from personal dosemeters. Radiat. Prot. Dosim. (2008) 129:469-472.

Belinsky SA, et al. Gene promoter methylation in plasma and sputum increases with lung cancer risk. Clinical cancer research : an official journal of the American Association for Cancer Research (2005) 11:6505-6511.

Blanchardon E, et al. Dosimetric protocol (CURE internal deliverable D2.2). DoReMi - Low Dose Research towards Multidisciplinary Integration - Task 5.8 “CURE project”. (2014a).

Blanchardon E, et al. Report on the evaluation of monitoring data and physico-chemical characterisation of radionuclides (CURE internal deliverable D2.1). DoReMi - Low Dose Research towards Multidisciplinary Integration - Task 5.8 “CURE project”. (2014b).

Boice JD, Jr., Cohen SS, Mumma MT, Chadda B, Blot WJ. A cohort study of uranium millers and miners of Grants, New Mexico, 1979-2005. Journal of radiological protection : official journal of the Society for Radiological Protection (2008) 28:303-325.

Boice JD, Jr., et al. Updated mortality analysis of radiation workers at Rocketdyne (Atomics International), 1948-2008. Radiation research (2011) 176:244-258.

Boice JD, Jr., et al. A comprehensive dose reconstruction methodology for former rocketdyne/atomics international radiation workers. Health physics (2006) 90:409-430.

Bonvallot N, et al. Metabolomics tools for describing complex pesticide exposure in pregnant women in Brittany (France). PloS one (2013) 8:e64433.

Brandom WF, Saccomanno G, Archer VE, Archer PG, Coors ME. Chromosome aberrations in uranium miners occupationally exposed to 222 radon. Radiation research (1972) 52:204-215.

Canu IG, Ellis ED, Tirmarche M. Cancer risk in nuclear workers occupationally exposed to uranium-emphasis on internal exposure. Health physics (2008) 94:1-17.

Canu IG, Laurent O, Pires N, Laurier D, Dublineau I. Health effects of naturally radioactive water ingestion: the need for enhanced studies. Environmental health perspectives (2011) 119:1676-1680.

Cardis E, et al. Risk of cancer after low doses of ionising radiation: retrospective cohort study in 15 countries. BMJ (2005) 331:77.

64

Carpenter L, Higgins C, Douglas A, Fraser P, Beral V, Smith P. Combined analysis of mortality in three United Kingdom nuclear industry workforces, 1946-1988. Radiation research (1994) 138:224-238.

Castellani CM, et al. IDEAS Guidelines (Version 2) for the Estimation of Committed Doses from Incorporation Monitoring Data. EURADOS Report 2013-01, Braunschweig, March 2013. (2013).

Chan C, et al. Mortality patterns among Paducah Gaseous Diffusion Plant workers. Journal of occupational and environmental medicine / American College of Occupational and Environmental Medicine (2010) 52:725-732.

Clarkson TW, Kench JE. Urinary excretion of amino acids by men absorbing heavy metals. Biochem J (1952) 62:361-371.

Currie G, Delles C. Proteinuria and its relation to cardiovascular disease. International journal of nephrology and renovascular disease (2013) 7:13-24.

Darolles C, et al. Different genotoxic profiles between depleted and enriched uranium. Toxicology letters (2010) 192:337-348.

Davesne E, Casanova P, Chojnacki E, Paquet F, Blanchardon E. Integration of uncertainties into internal contamination monitoring. Health physics (2010) 99:517-522.

Dounce AL, Roberts E, Wills JH. Catalasuria as a sensitive test for uranium poisoning. in : Voegtlin C, Hodge HC, ed. Pharmacology and toxicology of uranium compounds. Volume I. McGraw Hill, New York, NY 1949, pp 889-950. (1949).

Duport P, Robertson R, Ho K, Horvath F. Flow-through dissolution of uranium-thorium ore dust, uranium concentrate, uranium dioxide, and thorium alloy in simulated lung fluid. Radiat. Prot. Dosim. (1991) 38:121-133.

Dupree-Ellis E, Watkins J, Ingle JN, Phillips J. External radiation exposure and mortality in a cohort of uranium processing workers. American journal of epidemiology (2000) 152:91-95.

Dupree EA, Watkins JP, Ingle JN, Wallace PW, West CM, Tankersley WG. Uranium dust exposure and lung cancer risk in four uranium processing operations. Epidemiology (1995) 6:370-375.

Eisenbud M, Quigley JA. Industrial hygiene of uranium processing. A.M.A. archives of industrial health (1956) 14:12-22.

Elliott P, Peakman TC. The UK Biobank sample handling and storage protocol for the collection, processing and archiving of human blood and urine. International journal of epidemiology (2008) 37:234-244.

Engels H, et al. Radiation exposure and cause specific mortality among nuclear workers in Belgium (1969-1994). Radiation protection dosimetry (2005) 117:373-381.

Figgs LW. Lung cancer mortality among uranium gaseous diffusion plant workers: a cohort study 1952-2004. The international journal of occupational and environmental medicine (2013) 4:128-140.

Folstein MF, Folstein SE, McHugh PR. "Mini-mental state". A practical method for grading the cognitive state of patients for the clinician. Journal of Psychiatric Research (1975.) 12:189–198.

Garsi J-P, et al. Half-century archives of occupational medical data on French nuclear workers: a dusty warehouse or gold mine for epidemiological research? Arhiv za higijenu rada i toksikologiju (2014) 65.

Gillies M, Haylock R. The cancer mortality and incidence experience of workers at British Nuclear Fuels plc, 1946-2005. Journal of radiological protection : official journal of the Society for Radiological Protection (2014) 34:595-623.

Gilman AP, et al. Uranyl nitrate: 28-day and 91-day toxicity studies in the Sprague-Dawley rat. Toxicological sciences : an official journal of the Society of Toxicology (1998) 41:117-128.

Giussani A, et al. Final Report of the Uncertainties Working Group (UWG). (2014). Gomolka M, et al. The German Uranium Miners Biobank – current status and perspectives in

radiation research. 4th International MELODI Workshop. 12-14 September 2012, Helsinki, Finland (2012).

65

Gomolka M, et al. CURE Biology Protocol (CURE internal deliverable D3.2). DoReMi - Low Dose Research towards Multidisciplinary Integration - Task 5.8 “CURE project”. (2014).

Grignard E, Gueguen Y, Grison S, Lobaccaro JM, Gourmelon P, Souidi M. Contamination with depleted or enriched uranium differently affects steroidogenesis metabolism in rat. International journal of toxicology (2008) 27:323-328.

Grison S, et al. Metabolomics identifies a biological response to chronic low-dose natural uranium contamination in urine samples. Metabolomics : Official journal of the Metabolomic Society (2013) 9:1168-1180.

Grison S, et al. The metabolomic approach identifies a biological signature of low-dose chronic exposure to cesium 137. Journal of radiation research (2012) 53:33-43.

Gueguen Y, Rouas C. [New data on uranium nephrotoxicity]. Radioprotection (2012) 47. Guseva Canu I, Faust S, Knieczak E, Carles M, Samson E, Laurier D. Estimating historic exposures at

the European Gaseous Diffusion plants. International journal of hygiene and environmental health (2013) 216:499-507.

Guseva Canu I, et al. Does uranium induce circulatory diseases? First results from a French cohort of uranium workers. Occupational and environmental medicine (2012) 69:404-409.

Guseva Canu I, et al. Uranium carcinogenicity in humans might depend on the physical and chemical nature of uranium and its isotopic composition: results from pilot epidemiological study of French nuclear workers. Cancer causes & control : CCC (2011) 22:1563-1573.

Guseva Canu I, et al. Comparative assessing for radiological, chemical, and physical exposures at the French uranium conversion plant: Is uranium the only stressor? International journal of hygiene and environmental health (2009) 212:398-413.

Haylock R, Kreuzer M, Laurent O, Samson E. CURE Epidemiology Intermediate report (CURE internal deliverable D1.1). DoReMi - Low Dose Research towards Multidisciplinary Integration - Task 5.8 “CURE project”. (2014a).

Haylock R, Kreuzer M, Laurent O, Tomasek L, Engels H. CURE Epidemiology protocol (CURE internal deliverable D1.2). DoReMi - Low Dose Research towards Multidisciplinary Integration - Task 5.8 “CURE project”. (2014b).

Henny J, Zins M, Goldberg M, Dagher G, Clement B. Guide pour la constitution d’une biobanque associée aux études épidémiologiques en population générale. (2013) Paris: Lavoisier.

HLEG. High Level and Expert Group (HLEG). HLEG Report on European Low Dose Risk Research. EUR 23884. Luxembourg, UK: Office for Official Publications of the European Communities; 2009. (2009).

Hoffmann S, Guihenneuc C, Ancelet S. A Bayesian hierarchical approach to deal with exposure measurement error in the analysis of the cancer risk associated with exposure to ionizing radiation. An application to the French cohort of uranium miners. Journée Nationale du groupe Biopharmacie et Santé de la Société Française de Statistique. November 2014, Paris. (2014).

Houpert P, Lestaevel P, Bussy C, Paquet F, Gourmelon P. Enriched but not depleted uranium affects central nervous system in long-term exposed rat. Neurotoxicology (2005) 26:1015-1020.

IARC. IARC Monographs on the evaluation of carcinogenic risks to humans. Tobacco Smoke and involuntary smoking. (2004) 83.

ICRP. Protection against radon-222 at home and work. ICRP Publication 65. . Ann. ICRP (1993) 23. ICRP. Human respiratory tract model for radiological protection. ICRP Publication 66. Ann. ICRP

(1994) 24. ICRP. Age-dependent doses to members of the public from intakes of radionuclides: Part 3. Ingestion

dose coefficients. ICRP Publication 69. . Ann. ICRP (1995a) 25. ICRP. Dose coefficients for intakes of radionuclides by workers. Replacement of ICRP Publication 61.

ICRP Publication 68. Ann. ICRP (1995b) 24. ICRP. Human alimentary tract model for radiological protection. ICRP Publication 100. Ann. ICRP

(2006) 36.

66

ICRP. The 2007 Recommendations of the International Commission on Radiological Protection. ICRP Publication 103. Ann. ICRP 37 (2-4). (2007).

ICRP. Nuclear decay data for dosimetric calculations. ICRP Publication 107. Ann. ICRP (2008) 38. ICRP. Adult reference computational phantoms. ICRP Publication 110. Ann. ICRP (2009) 39. ICRP. Occupational Intakes of Radionuclides, Part 1. Annals of the International Commission on

Radiological Protection, Accepted for publication. (2015a). ICRP. Occupational Intakes of Radionuclides, Part 2. Annals of the International Commission on

Radiological Protection, Accepted for publication. (2015b). ICRP. Occupational Intakes of Radionuclides, Part 3. Annals of the International Commission on

Radiological Protection, Accepted for publication. (2015c). ICRP. Occupational Intakes of Radionuclides, Part 4. Annals of the International Commission on

Radiological Protection, Accepted for publication. (2015d). ICRP. Values of specific absorbed fractions (provisional title). In preparation. (2015e). ICRU. Fundamental Quantities and Units for Ionizing Radiation (Report 60). (1993). Johnson P, Atkinson WD, Nicholls JL. Updated analysis of mortality in workers at UK atomic weapons

establishments. Proceedings of the 6th International SRP symposium. SRP, London. (1999). Kazymbet P. Radiobiological and Radioecological Research in Uranium Mining Regions of Kazakhstan.

Presented at the Fifth International Scientific and Practical Conference "Biomedical and radioecological problems in the uranium mining regions" June 19 (2014) Astana, Kazakhstan.

Kreuzer M, et al. Mortality from internal and external radiation exposure in a cohort of male German uranium millers, 1946-2008. International archives of occupational and environmental health (2014).

Kreuzer M, Kreisheimer M, Kandel M, Schnelzer M, Tschense A, Grosche B. Mortality from cardiovascular diseases in the German uranium miners cohort study, 1946-1998. Radiation and environmental biophysics (2006) 45:159-166.

Kreuzer M, Schnelzer M, Tschense A, Walsh L, Grosche B. Cohort profile: the German uranium miners cohort study (WISMUT cohort), 1946-2003. International journal of epidemiology (2010) 39:980-987.

Kreuzer M, et al. Silica dust, radon and death from non-malignant respiratory diseases in German uranium miners. Occupational and environmental medicine (2013) 70:869-875.

Krunic A, Haveric S, Ibrulj S. Micronuclei frequencies in peripheral blood lymphocytes of individuals exposed to depleted uranium. Arhiv za higijenu rada i toksikologiju (2005) 56:227-232.

Kurttio P, et al. Renal effects of uranium in drinking water. Environmental health perspectives (2002) 110:337-342.

Kurttio P, et al. Kidney toxicity of ingested uranium from drinking water. American journal of kidney diseases : the official journal of the National Kidney Foundation (2006) 47:972-982.

Kurttio P, Komulainen H, Leino A, Salonen L, Auvinen A, Saha H. Bone as a possible target of chemical toxicity of natural uranium in drinking water. Environmental health perspectives (2005) 113:68-72.

Laurier D, et al. DoReMi workshop on multidisciplinary approaches to evaluating cancer risks associated with low-dose internal contamination. Radioprotection (2012) 47:119-148.

Laurier D, et al. Quantification of cancer and non-cancer risks associated with multiple chronic radiation exposures: Epidemiological studies, organ dose calculation and risk assessment. Alpha-Risk project deliverable D1.8. Analysis of risk among miners using organ dose calculation. Funded by the European Commission EC FP6 (Ref. FI6R-CT-2005-516483). European Commission DG XII, Brussels. (2009).

Leach LJ, Yuile CL, Hodge HC, Sylvester GE, Wilson HB. A five-year inhalation study with natural uranium dioxide (UO2) dust. II. Postexposure retention and biologic effects in the monkey, dog and rat. Health physics (1973) 25:239-258.

Leggett R, Marsh J, Gregoratto D, Blanchardon E. A generic biokinetic model for noble gases with application to radon. Journal of radiological protection : official journal of the Society for Radiological Protection (2013) 33:413-432.

67

Leng S, et al. Genetic variation in SIRT1 affects susceptibility of lung squamous cell carcinomas in former uranium miners from the Colorado plateau. Carcinogenesis (2013) 34:1044-1050.

Lestaevel P, Houpert P, Bussy C, Dhieux B, Gourmelon P, Paquet F. The brain is a target organ after acute exposure to depleted uranium. Toxicology (2005) 212:219-226.

Leuraud K, et al. Radon, smoking and lung cancer risk: results of a joint analysis of three European case-control studies among uranium miners. Radiation research (2011) 176:375-387.

Li K, et al. Alteration of cytokine profiles in uranium miners exposed to long-term low dose ionizing radiation. TheScientificWorldJournal (2014) 2014:216408.

Li WB, Janzen T, Zankl M, Giussani A, Hoeschen C. Uncertainties of organ-absorbed doses to patients from 18f-choline. Proc. SPIE 7961, Medical Imaging 2011: Physics of Medical Imaging, 796129 (March 16, 2011). (2011).

Luessenhop AJ, Gallimore JC, Sweet WH, Struxness EG, Robinson J. The toxicity in man of hexavalent uranium following intravenous administration. The American journal of roentgenology, radium therapy, and nuclear medicine (1958) 79:83-100.

Mao Y, et al. Inorganic components of drinking water and microalbuminuria. Environmental research (1995) 71:135-140.

Marsh JW, et al. Dosimetric calculations for uranium miners exposed to radon gas, radon progeny and long-lived radionuclides. Alpha-Risk project deliverables D5.5 & D5.6 Lung dosimetry model for uranium dust inhalation; organ dosimetry for radon progeny and long-lived radionuclides. Funded by the European Commission EC FP6 (Ref. FI6R-CT-2005-516483). European Commission DG XII, Brussels. (2008).

Marsh JW, et al. Alpha Miner Software Program (version 4.0). Alpha-Risk project deliverable D5.9. Final analysis of dose calculations and risk assessment for dose-response modelling. Funded by the European Commission EC FP6 (Ref. FI6R-CT-2005-516483). European Commission DG XII, Brussels. . (2009).

Marsh JW, et al. Evaluation of scattering factor values for internal dose assessment following the IDEAS guidelines: preliminary results. Radiation protection dosimetry (2007) 127:339-342.

Marsh JW, et al. Dosimetric calculations for uranium miners for epidemiological studies. Radiation protection dosimetry (2012) 149:371-383.

Martin F, Earl R, Tawn EJ. A cytogenetic study of men occupationally exposed to uranium. British journal of industrial medicine (1991) 48:98-102.

McDiarmid MA, et al. Measures of genotoxicity in Gulf war I veterans exposed to depleted uranium. Environmental and molecular mutagenesis (2011a) 52:569-581.

McDiarmid MA, et al. Longitudinal health surveillance in a cohort of Gulf War veterans 18 years after first exposure to depleted uranium. Journal of toxicology and environmental health. Part A (2011b) 74:678-691.

McDiarmid MA, et al. The Gulf War depleted uranium cohort at 20 years: bioassay results and novel approaches to fragment surveillance. Health physics (2013) 104:347-361.

McGeoghegan D, Binks K. The mortality and cancer morbidity experience of workers at the Capenhurst uranium enrichment facility 1946-95. Journal of radiological protection : official journal of the Society for Radiological Protection (2000a) 20:381-401.

McGeoghegan D, Binks K. The mortality and cancer morbidity experience of workers at the Springfields uranium production facility, 1946-95. Journal of radiological protection : official journal of the Society for Radiological Protection (2000b) 20:111-137.

Milacic S. Health investigations of depleted-uranium clean-up workers. La Medicina del lavoro (2008) 99:366-370.

Milacic S, Simic J. Identification of health risks in workers staying and working on the terrains contaminated with depleted uranium. Journal of radiation research (2009) 50:213-222.

Mitchel RE, Jackson JS, Heinmiller B. Inhaled uranium ore dust and lung cancer risk in rats. Health physics (1999) 76:145-155.

68

Monleau M, De Meo M, Paquet F, Chazel V, Dumenil G, Donnadieu-Claraz M. Genotoxic and inflammatory effects of depleted uranium particles inhaled by rats. Toxicological sciences : an official journal of the Society of Toxicology (2006) 89:287-295.

NCRP. Development of a biokinetic model for radionuclide-contaminated wounds and procedures for their assessment, dosimetry and treatment. National Council on Radiation Protection and Measurements Report No. 156, NCRP: Bethesda, USA. (2006).

NRC. National Research Council. Comparative dosimetry of radon in mines and homes. National Academy Press (Washington, DC). ISBN 0-309-04484-7. (1991).

NRC. National Research Council, Committee on Health Risks of Exposure to Radon. Health effects of exposure to radon (BEIR VI). Washington, DC: National Academy Press. (1999).

NRC. National Research Council. Health risks from exposures to low levels of ionizing radiation (BEIR VII), phase 2. Board on radiation effects research. National Academies Press, Washington. (2006).

Palmer LJ. UK Biobank: bank on it. Lancet (2007) 369:1980-1982. Pernot E, Cardis E, Badie C. Usefulness of saliva samples for biomarker studies in radiation research.

Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology (2014) 23:2673-2680.

Pernot E, et al. Ionizing radiation biomarkers for potential use in epidemiological studies. Mutation research (2012) 751:258-286.

Pinkerton LE, Bloom TF, Hein MJ, Ward EM. Mortality among a cohort of uranium mill workers: an update. Occupational and environmental medicine (2004) 61:57-64.

Poisson C, et al. Chronic uranium exposure dose-dependently induces glutathione in rats without any nephrotoxicity. Free radical research (2014) 48:1218-1231.

Prat O, et al. From cell to man: evaluation of osteopontin as a possible biomarker of uranium exposure. Environment international (2011) 37:657-662.

Puncher M, Birchall A, Bull RK. A Bayesian analysis of uncertainties on lung doses resulting from occupational exposures to uranium. Radiation protection dosimetry (2013) 156:131-140.

Rage E, Caer-Lorho S, Drubay D, Ancelet S, Laroche P, Laurier D. Mortality analyses in the updated French cohort of uranium miners (1946-2007). International archives of occupational and environmental health (2014).

Rage E, et al. Risk of lung cancer mortality in relation to lung doses among French uranium miners: follow-up 1956-1999. Radiation research (2012) 177:288-297.

Richardson DB, Wing S. Lung cancer mortality among workers at a nuclear materials fabrication plant. American journal of industrial medicine (2006) 49:102-111.

Riddell AE. Uranium exposure assessment - a critical review. BNES - Radiation dose management. (1995).

Riddell AE. An analysis of the Impact of Aerosol Lung Solubility Assumptions on Dose Estimates. Proc. of the Seventh Internal Symposium of the Society for Radiological Protection, Cardiff. (2005).

Ritz B. Cancer mortality among workers exposed to chemicals during uranium processing. Journal of occupational and environmental medicine / American College of Occupational and Environmental Medicine (1999a) 41:556-566.

Ritz B. Radiation exposure and cancer mortality in uranium processing workers. Epidemiology (1999b) 10:531-538.

Samson E, et al. Mortality from cancer and non cancer diseases in the French cohort of uranium processing workers (TRACY). (in preparation).

Samson E, Piot I, Zhivin S, Acker A, Laroche P, Laurier D. Mortality profile of the French cohort of uranium processing workers. Occupational and environmental medicine (2014) 71 Suppl 1:A23.

Schlesinger RB, Kunzli N, Hidy GM, Gotschi T, Jerrett M. The health relevance of ambient particulate matter characteristics: coherence of toxicological and epidemiological inferences. Inhalation toxicology (2006) 18:95-125.

69

Selden AI, et al. Nephrotoxicity of uranium in drinking water from private drilled wells. Environmental research (2009) 109:486-494.

Silver SR, et al. Mortality and ionising radiation exposures among workers employed at the Fernald Feed Materials Production Center (1951-1985). Occupational and environmental medicine (2013) 70:453-463.

Stearns DM, et al. Uranyl acetate induces hprt mutations and uranium-DNA adducts in Chinese hamster ovary EM9 cells. Mutagenesis (2005) 20:417-423.

Takhauov RM, et al. The Bank of Biological Samples Representing Individuals Exposed to Long-Term Ionizing Radiation at Various Doses. Biopreservation and biobanking (2015).

Thun MJ, Baker DB, Steenland K, Smith AB, Halperin W, Berl T. Renal toxicity in uranium mill workers. Scandinavian journal of work, environment & health (1985) 11:83-90.

Tomasek L. Lung cancer mortality among Czech uranium miners-60 years since exposure. Journal of radiological protection : official journal of the Society for Radiological Protection (2012) 32:301-314.

Tomasek L. Effect of age at exposure in 11 underground miners studies. Radiation protection dosimetry (2014) 60:124-127.

UNSCEAR. United Nations Scientific Committee on the Effects of Atomic Radiation. UNSCEAR 2006 Report to the General Assembly with Scientific Annexes, Effects of Ionizing Radiation. Vol. 1: Report and Annexes A and B New York, NY: United Nations. (2008a).

UNSCEAR. United Nations Scientific Committee on the Effects of Atomic Radiation. UNSCEAR 2006. Sources and effects of Ionizing Radiation. Annex B. Exposure of the public and workers from various sources of radiation. (2008b).

UNSCEAR. United Nations Scientific Committee on the Effects of Atomic Radiation. Sources, effects and risks of ionizing radiation. UNSCEAR 2012 Report to the General Assembly. Scientific annexes A and B. Annex B. Uncertainties in risk estimates for radiation-induced cancer. New York, NY: United Nations. (2012).

Wiethege T, et al. German uranium miner study--pathological and molecular genetic findings. German Uranium Miner Study, Research Group Pathology. Radiation research (1999) 152:S52-55.

Yiin JH, et al. A nested case-control study of multiple myeloma risk and uranium exposure among workers at the Oak Ridge Gaseous Diffusion Plant. Radiation research (2009) 171:637-645.

Zablotska LB, Lane RS, Frost SE. Mortality (1950-1999) and cancer incidence (1969-1999) of workers in the Port Hope cohort study exposed to a unique combination of radium, uranium and gamma-ray doses. BMJ open (2013) 3.

Zamora ML, Zielinski JM, Moodie GB, Falcomer RA, Hunt WC, Capello K. Uranium in drinking water: renal effects of long-term ingestion by an aboriginal community. Archives of environmental & occupational health (2009) 64:228-241.

Zhivin S, Laurier D, Guseva Canu I. Health effects of occupational exposure to uranium: do physicochemical properties matter? International journal of radiation biology (2014) 11:1-33.

Zhu G, Xiang X, Chen X, Wang L, Hu H, Weng S. Renal dysfunction induced by long-term exposure to depleted uranium in rats. Archives of toxicology (2009) 83:37-46.

Zins M, et al. The CONSTANCES cohort: an open epidemiological laboratory. BMC public health (2010) 10:479.

Zolzer F, et al. Persistence of genetic damage in lymphocytes from former uranium miners. Cytogenetic and genome research (2012a) 136:288-294.

Zolzer F, et al. Micronuclei in lymphocytes from currently active uranium miners. Radiation and environmental biophysics (2012b) 51:277-282.

70

LIST OF PARTICIPANTS TO THE CURE PROJECT

Sophie Ancelet, IRSN Will Atkinson, NUVIA Ltd Sarah Baatout, SCK•CEN Christophe Badie, PHE Gary Bethel, NUVIA Ltd Jean-Marc Bertho, IRSN Derek Bingham, AWE Eric Blanchardon, IRSN Richard Bull, NUVIA Limited Elisabeth Cardis, CREAL Cécile Challeton De Vathaire, IRSN Rupert Cockerill, AWE Estelle Davesne, IRSN Damien Drubay*, IRSN Teni Ebrahimian, IRSN Hilde Engels, SCK•CEN Nora Fenske, BfS Michael Gillies, PHE Augusto Giussani, BfS Maria Gomolka, BfS James Grellier, CREAL Stephane Grison, IRSN Yann Gueguen, IRSN Janet Hall, IC Richard Haylock, PHE Sabine Hornhardt, BfS Chrystelle Ibanez, IRSN Lukas Kotik, SURO Michaela Kreuzer, BfS Olivier Laurent, IRSN Dominique Laurier, IRSN Anne-Laure Le Bacq, SCK•CEN James Marsh, PHE Dietmar Nosske, BfS Jackie O'hagan, PHE Eileen Pernot , CREAL Matthew Puncher, PHE Roel Quintens, SCK•CEN Estelle Rage, IRSN Tony Riddell, PHE Laurence Roy, IRSN Eric Samson, IRSN Maamar Souidi, IRSN Ladislav Tomasek, SURO Michelle Turner, CREAL Nina Weiland, BfS Sergey Zhivin*, IRSN

*PhD student

71

LIST OF INVITED EXPERTS

Naomi Allen, University of Oxford (UK)

Jeri Anderson, National Institute for Occupational Safety and Health (USA)

Meirat Batkin, Astana Medical University (Kazakhstan)

Steven Belinsky, Lovelace Respiratory Research Institute (USA)

Georges Dagher, Inserm (France)

Andrey Karpov, Seversk Biophysical Research Center (Russia)

Polat Kazymbet, Astana Medical University (Kazakhstan)

Richard Wakeford, University of Manchester (UK)

Friedo Zölzer, University of South Bohemia (Czech Republic)

Petr Otahal, National Institute for Nuclear, Chemical and Biological Protection (Czech Republic)

72

GLOSSARY ADU: ammonium diuranate AMAD: activity median aerodynamic diameter AWE: Atomic Weapons Establishment BBMRI: Biobanking and Biomolecular Resources Research Infrastructure BfS: Bundesamt für Strahlenschutz CRA: clinical research associate CREAL: Centre de Recerca en Epidemiologia Ambiental CURE: Concerted Uranium Research in Europe EDTA: Ethylenediaminetetraacetic acid HRTM: human respiratory tract model HATM: human alimentary tract model IC: Institut Curie IRSN : Institut de Radioprotection et de Sûreté Nucléaire JCCRER: Joint Coordinating Committee for Radiation Effects Research JEM: job exposure matrix LD: limit of detection LLR: Long Lived Radionuclides LSS: Life Span Study MOX: mixed oxide fuels PHE: Public Health England RL: reporting limit SF: scattering factor SMR: Standardised Mortality Ratios (SMRs) SCK•CEN: StudieCentrum voor Kernenergie • Centre d'étude de l'Energie Nucléaire SOP: standard operating procedure SURO: Státní ústav radiační ochrany TBP: tributyl phosphate UKAEA: United Kingdom Atomic Energy Authority WL: working level WLM: working level month WP: work package WR: radiation weighting factor

73

ACKNOWLEDGEMENTS The CURE project was supported by the EU FP7 DoReMi Network of Excellence (grant agreement:

249689)

74

ANNEXES

Annex 1. Simplified scheme of the nuclear fuel cycle

Annex 2. Early dissemination activities

75

Annex 1. Simplified scheme of the nuclear fuel cycle.

76

Annex 2. Early dissemination activities Oral presentations: Laurier D, Gomolka M, Haylock R, Atkinson W, Bingham D, Baatout S, Tomasek L, Cardis E, Hall J,

Blanchardon E. Concerted Action for an Integrated (biology-dosimetry-epidemiology) Research

project on Occupational Uranium Exposure. Fifth International MELODI Workshop, Bruxelles

(Belgium), Oct 2013.

Laurier D, Gomolka M, Haylock R, Atkinson W, Bingham D, Baatout S, Tomasek L, Cardis E, Hall J,

Blanchardon E. The CURE project: a Concerted Action for an Integrated (biology-dosimetry-

epidemiology) Research project on Occupational Uranium Exposure. Fourth congress of International

Radiation Protection Association (IRPA), Genova (Switzerland), June 24, 2014.

Laurent O, Gomolka M, Haylock R, Atkinson W, Bingham D, Baatout S, Tomasek L, Cardis E, Hall J,

Blanchardon E, Laurier D. Concerted Uranium Research in Europe: the CURE project. Conference

"Biomedical and radio-ecological problems in the uranium mining regions". Astana (Kazakhstan),

June 19, 2014.

Laurier D. Progress of the CURE project. 3rd DoReMi periodic meeting. Munich (Germany), July 9,

2014

Laurier D, Gomolka M, Haylock R, Atkinson W, Bingham D, Baatout S, Tomasek L, Cardis E, Hall J,

Blanchardon E. Concerted Action for an Integrated (biology dosimetry-epidemiology) Research

Project on Occupational Uranium Exposure. 3rd DoReMi periodic meeting. Munich (Germany), July 9,

2014

Poster presentations:

Laurent O, Gomolka M, Haylock R, Giussani A, Atkinson W, Bingham D, Baatout S, Tomasek L, Cardis

E, Hall J, Blanchardon E, Laurier D. Concerted Uranium Research In Europe (CURE): Recent Progresses

And Perspectives. MELODI annual workshop, Barcelona (Spain), October 7-9 2014

CURE deliverables:

Laurier D, Roy L, Haylock R, Rage E, Blanchardon E, Giussani A, Gomolka M, Bertho J-M. DoReMi - Low Dose Research towards Multidisciplinary Integration Task 5.8 “CURE project”CURE. Kick-off meeting report (CURE internal deliverable 4.1), November 2013. Haylock R, Kreuzer M, Laurent O, Samson E. DoReMi - Low Dose Research towards Multidisciplinary Integration Task 5.8 “CURE project”. CURE Epidemiology Intermediate report (WP1) (CURE internal Deliverable D1.1), May 2014. Banchardon, E, Bingham D, Bull R, Challeton - de Vathaire C, Davesne E, Giussani A, Hochstrat B, Kreuzer M, Le Bacq A.-L., Noßke D, Marsh J, Rage E, Riddell T, Samson E, Tomášek L, Tschense A, Zhivin S. DoReMi - Low Dose Research towards Multidisciplinary Integration Task 5.8 “CURE project”. Report on the evaluation of monitoring data and physico-chemical characterisation of radionuclides (WP2) (CURE internal Deliverable D2.1), May 2014.

77

Gomolka M, Badie C, Baatout S, Bertho J-M, Ebrahimian T, El Saghire H, Gueguen Y, Hall J, Hornhardt S, Ibanez C, Kabacik S, Pernot E, Roy L. DoReMi - Low Dose Research towards Multidisciplinary Integration Task 5.8 “CURE project”. Biology intermediary report (WP3) (CURE internal Deliverable D3.1), May 2014. Haylock R, Kreuzer M, Laurent O, Tomášek L, Engels H. DoReMi - Low Dose Research towards Multidisciplinary Integration Task 5.8 “CURE project”. Epidemiology protocol (WP1) (CURE deliverable D1.2), December 2014. Blanchardon E, Bingham D, Bull R, Challeton - de Vathaire C, Cockerill R, Davesne E, Giussani A, Le Bacq A.-L., Noßke D, Marsh J, Riddell T, Tomášek L. DoReMi - Low Dose Research towards Multidisciplinary Integration Task 5.8 “CURE project”. Dosimetric protocol (WP2) (CURE deliverable D2.2), December 2014. Gomolka M, Hornhardt S, Weiland N, Roy L, Bertho J-M, Gueguen Y, Ibanez C, Ebrahimian T, Grison S, Souidi M, Hall J, Badie C, Kabacik S, Pernot E, Turner M, Baatout S, Quintens R, Zölzer F, Belinsky S. DoReMi - Low Dose Research towards Multidisciplinary Integration Task 5.8 “CURE project”. Biology Protocol (WP3) (CURE deliverable D3.2), December 2014. Giussani A, Ancelet S, Atkinson W, Bethel G, Bingham D, Blanchardon E, Bull, R, Cockerill R, Davesne E, Fenske N, Grellier J, Hall J, Haylock R, Hornhardt S, Kotik L, Laurent O, Laurier D, Marsh J, Nosske D, Puncher M, Rage E, Riddell T, Samaga D, Tomasek L. DoReMi - Low Dose Research towards Multidisciplinary Integration Task 5.8 “CURE project”. Final Report of the Uncertainties Working Group (UWG), December 2014.


Recommended