Beskæftigelsesudvalget 2019-20
BEU Alm.del Bilag 101
Offentligt
2130137_0001.png
Titanium
dioxide
nanomaterials:
Scientific basis
for setting
a health-based
occupational
exposure limit
Anne Thoustrup Saber, Sarah Søs Poulsen, Niels Hadrup,
Karin Sørig Hougaard, Nicklas Raun Jacobsen and Ulla Vogel
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
TITANIUM DIOXIDE NANOMATERIALS:
SCIENTIFIC BASIS FOR SETTING A HEALTH-
BASED OCCUPATIONAL EXPOSURE LIMIT
Anne Thoustrup Saber
Sarah Søs Poulsen
Niels Hadrup
Karin Sørig Hougaard
Nicklas Raun Jacobsen
Ulla Vogel
The National Research Centre for the Working Environment, Copenhagen 2018
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
NFA-report
Title
Titanium dioxide nanomaterials: Scientific basis for setting a health-based
occupational exposure limit
Anne Thoustrup Saber, Sarah Søs Poulsen, Niels Hadrup, Karin Sørig
Hougaard, Nicklas Raun Jacobsen and Ulla Vogel
The National Research Centre for the Working Environment (NFA)
The National Research Centre for the Working Environment (NFA)
September 2018
978-87-7904-351-0
nfa.dk
Authors
Institution
Publisher
Published
ISBN
Internet version
The National Research Centre for the Working Environment (NFA)
Lersø Parkallé 105
DK-2100 Copenhagen
Phone: +45 39165200
Fax: +45 39165201
e-mail: [email protected]
Website: nfa.dk
ii
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
2130137_0005.png
F
OREWORD
In 2015, the Danish Working Environment Council made 22 recommendations to
promote safe handling of nanomaterials (NMs) in the working environment, which were
enforced by the Minister of Employment. One of these recommendations was ‘That the
Danish Working Environment Authority in cooperation with relevant scientific experts
assesses whether adequate scientific documentation can be provided to use the scientific
quality committee for an assessment of the scientific evidence to determine limit values
for specific NMs in the work environment.’ (https://www.amr.dk/nano.aspx).
On this background, The Danish Working Environment Authority asked NFAto review
the scientific evidence with the aim of clarifying the possibilities for suggesting
nanospecific occupational exposure limits (OELs) for three different NMs (titanium
dioxide (TiO
2
), carbon black and carbon nanotubes (CNT)).
The purpose of the present report is to suggest a health-based OEL for nanosized TiO
2
.
Elizabeth Bengtsen and Karen Bo Frydendall, NFA, are gratefully acknowledged for
assistance with literature search.
The working group wishes to thank Chief Toxicologist Poul Bo Larsen, DHI, Denmark,
for reviewing the report.
Copenhagen, August 2018
iii
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
E
XECUTIVE SUMMARY
In this report, a working group at the National Research Centre for the Working
Environment reviews data relevant to assessing the hazard of TiO
2
nanomaterials (TiO
2
NMs), i.e. human studies (Chapter 2), toxicokinetics (Chapter 3), animal studies (Chapter
4), mechanisms of toxicity (Chapter 5), previous risk assessments of TiO
2
NMs (Chapter
6), scientific basis for setting an occupational exposure limit (OEL) (Capter 7) and finally
we summarize and suggest a health based OEL for TiO
2
NM (Chapter 8). The focus of
this report is only occupational exposure by inhalation.
The present working group evaluated the relevant literature on TiO
2
NM from both
epidemiological and animal inhalation studies. None of the identified epidemiological
studies provided information on the particle size range of the TiO
2
, thus making it
impossible to determine whether the exposures included TiO
2
NM. Therefore it was
decided to base the suggested health-based OEL on data from experimental animal
studies.
Pulmonary inflammation and carcinogenicity was observed in inhalation studies in rats.
The present working group regards inflammation and carcinogenicity as the critical
adverse effects and the subsequent risk assessments are conducted based on studies
reporting these effects. TiO
2
NM induced cardiovascular effects were identified in animal
studies. However, none of these studies were sub-chronic or chronic inhalation studies
and therefore not suitable for OEL derivation. However, the present working group
regards the acute phase response as a biomarker of cardiovascular effects. Due to the
close association between pulmonary inflammation and the acute phase response, the
present working group regards inflammation as a proxy for cardiovascular effects
mediated by the acute phase response.
The present working group found strong dose response relationships for neutrophil
influx as a marker of pulmonary inflammation. Neutrophil influx was related to
deposited surface area. The working group considers inflammation as a threshold effect.
The present working group found that the mechanism of action of the carcinogenic effect
has not been fully clarified. Secondary genotoxicity due to particle-induced
inflammation is an important and well documented mechanism of action for the
development of lung cancer. However, the available data did not allow ruling out that
TiO
2
NM could also induce cancer through a direct genotoxic mechanism. Therefore, the
present working group considers carcinogenicity as a non-threshold effect.
Consequently, the present working group decided to perform the risk assessment based
on both a threshold effect for inflammation and a non-threshold effect for cancer.
For an OEL based on threshold effects, the following traditional approach suggested by
REACH is utilized: 1) identification of critical effect, 2) identification of the no observed
adverse effect concentration (NOAEC), 3) calculation of OEL using assessment factors
adjusting for inter and intra species differences. For non-threshold effects, the present
working group uses two approaches. The first method uses the measured lung burden in
rats exposed by inhalation and the alveolar surface area of rats and humans to estimate
iv
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
2130137_0007.png
the human equivalent lung burden. The second method, suggested by The European
Chemicals Agency (ECHA), uses air concentrations directly.
The working group considered that data from two rat inhalation studies as the best basis
for the risk assessment. The following studies were selected to be used for calculation of
the derived-no-effect level (DNEL) and dose-response for excess cancer risk,
respectively: A 13 week sub-chronic inhalation study in rats (0, 0.5, 2.0 and 10 mg/m
3
)
and a 2 year chronic cancer inhalation study in rats (0 and 10 mg/m
3
). The table below
shows a DNEL for pulmonary inflammation derived based on the sub-chronic inhalation
study of rats, and extra lung cancer risk at 1 in 1 000, 1 in 10 000 and 1 in 100 000 derived
using two different approaches.
Overview of DNEL based on a threshold based mechanism of action and exposure levels
resulting in extra cancer risk levels at 1:1000, 1:10 000 and 1: 100 000 based on a non-threshold
based mechanism of action.
Mechanism of
action
Threshold
based
Non-treshold
based
DNEL
Extra cancer
risk
1:1000
1:10 000
1:100 000
Suggestion of an OEL for TiO
2
NM
Inflammation
Lung cancer
Lung cancer
(method I)
(method II)
10 µg/m
3
4 µg/m
3
0.4 µg/m
3
0.04 µg/m
3
47 µg/m
3
4.7 µg/m
3
0.47 µg/m
3
Both studies used for the risk assessment used P25 TiO
2
NM (15-40 nm diameter, 80%
anatase/20% rutile). TiO
2
NMs differ regarding size and surface area but also coating,
shape, crystal structure etc. The present working group notes that there is limited
available data on the biological effects of different physico-chemical properties, but the
present working group concludes that the majority of available data support that the
surface area (and therefore the size) of TiO
2
is a critical driver of particle-induced
inflammation and the acute phase response in the lungs. In support of this notion, The
National Institute for Occupational Safety and Health (NIOSH) showed that the
deposited surface area of TiO
2
particles of different sizes (fine and ultrafine) and
different crystal structure (80% anatase/20% rutile and 99% rutile) can explain the
observed variation in TiO
2
particle-induced pulmonary inflammation and lung cancer in
rat inhalation studies. This stresses the importance of the surface area as a predictor for
the inflammatory and carcinogenic response.
The present working group regards cancer as the most critical effect. The DNEL
approach relies heavily on the assumption of a threshold effect on inflammation and
carcinogenicity. The present working group is of the opinion that there is still
uncertainly whether this is the case for TiO
2
NM–induced carcinogenicity.
Two different approaches were used for calculating excess lung cancer risk based on the
same chronic inhalation study in rats. In the first approach, lung burden was used to
v
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
estimate the exposure levels. In the second approach, air concentrations were used
directly. Independently of the applied method for risk assessment, the resulting OEL
suggestions were all very low. These levels are all more than 100-fold lower than the
current Danish OEL for titanium of 6 mg/m
3
(measured as Ti, corresponding to 10 mg/m
3
TiO
2
).
The present working group recommends the risk assessment approach estimating the
excess lung cancer risk based on lung burden, since this approach takes the retained
pulmonary dose into account. Thus, the expected excess lung cancer risk in relation to
occupational exposure to TiO
2
NMs is 1:1 000 at 4 µg/m
3
, 1:10 000 at 0.4 µg/m
3
and 1:100
000 at 0.04 µg/m
3
TiO
2
NM.
vi
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
D
ANSK SAMMENFATNING
I denne rapport vurderer en arbejdsgruppe ved Det Nationale Forskningscenter for
Arbejdsmiljø data, der er relevante for at vurdere faren ved udsættelse for titanium
dioxid nanomaterialer (TiO
2
NM), dvs. humane studier (kapitel 2), toksikokinetik
(kapitel 3), dyreforsøg (kapitel 4), toksicitetsmekanismer (kapitel 5), tidligere
risikovurderinger af TiO
2
NM (kapitel 6), det videnskabelige grundlag for fastlæggelse af
en grænseværdi (kapitel 7) og endelig opsummeres og foreslås en helbredsbaseret
grænseværdi for TiO
2
NM i arbejdsmiljøet (kapitel 8). Fokus i denne rapport er alene på
erhvervsmæssig eksponering ved indånding.
Den nærværende arbejdsgruppe evaluerede den relevante litteratur om TiO
2
NM fra
både epidemiologiske undersøgelser og inhalationsforsøg med dyr. Ingen af de
identificerede epidemiologiske undersøgelser indeholdt oplysninger om TiO
2
's
partikelstørrelse, hvilket gør det umuligt at afgøre, om eksponeringerne omfattede TiO
2
NM. Derfor blev det besluttet at basere den foreslåede sundhedsbaserede grænseværdi i
arbejdsmiljøet på data fra studier på forsøgsdyr.
Der blev observeret lungeinflammation og lungekræft i inhalationsundersøgelser af
rotter. Den nærværende arbejdsgruppe anser inflammation og kræft som de vigtigste
skadelige effekter. Derfor baseres de efterfølgende risikovurderinger på undersøgelser,
der rapporterer om disse effekter. Der blev identificeret TiO
2
NM-inducerede
kardiovaskulære effekter i dyreforsøg. Ingen af disse undersøgelser var imidlertid
subkroniske eller kroniske inhalationsundersøgelser og derfor var de ikke egnede til
risikovurdering. Den nærværende arbejdsgruppe anser dog akutfaseresponset som en
biomarkør for kardiovaskulære effekter. På grund af den stærke sammenhæng mellem
lungeinflammation og akutfasesponset betragter den nærværende arbejdsgruppe
inflammation som en proxy for hjertekareffekter medieret af akutfaserespons.
Den nærværende arbejdsgruppe fandt stærk dosis-respons-sammenhæng for neutrofilt
influx som markør for lungeinflammation. Det samlede lungedeponerede specifikke
overfladeareal prædikterede neutrofilt influx. Arbejdsgruppen anser inflammation for at
være en tærskeleffekt.
Den nærværende arbejdsgruppe fandt, at virkningsmekanismen for den
kræftfremkaldende effekt ikke er blevet fuldstændigt afklaret. Sekundær genotoksicitet
forårsaget af partikelinduceret inflammation er en vigtig og veldokumenteret
virkningsmekanisme for udvikling af lungekræft. De tilgængelige data tillod dog ikke at
udelukke at TiO
2
NM også kunne inducere kræft gennem en direkte genotoksisk
mekanisme. Derfor anser den nærværende arbejdsgruppe kræft som ikke-tærskel effekt.
Det blev derfor besluttet at udføre risikovurderingen baseret på både en tærskeleffekt for
inflammation og en ikke-tærskeleffekt for kræft.
For en grænseværdi i arbejdsmiljøet baseret på tærskeleffekt anvendes følgende
traditionelle tilgang, som anbefalet af REACH: 1) identifikation af kritisk effekt, 2)
identifikation af NOAEC, og 3) beregning af grænseværdi ved anvendelse af
vurderingsfaktorer, der justerer for inter- og intraspecies forskelle. For ikke-
vii
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
2130137_0010.png
tærskeleffekter anvender den nærværende arbejdsgruppe to metoder. Ved den første
metode anvendes den målte lungedeponerede dosis hos rotter til at estimere den
tilsvarende eksponering i arbejdsmiljøet. Ved den anden metode anvendes
luftkoncentrationer direkte.
Arbejdsgruppen fandt, at data fra to inhalationsundersøgelser i rotter var det bedste
grundlag for risikovurderingen. Følgende undersøgelser blev udvalgt til beregning af
henholdsvis DNEL og kræftrisiko: En 13-ugers subkronisk inhalationsundersøgelse af
rotter (0, 0,5, 2,0 og 10 mg/m
3
) og en 2-årig kronisk kræftinhalationsundersøgelse af
rotter (0 og 10 mg/m
3
). Tabellen nedenfor viser en DNEL for lungeinflammation beregnet
på basis af det subkroniske inhalationsstudie af rotter og ekstra lungekræftrisiko hos 1
ud af 1.000, 1 ud af 10.000 og 1 ud af 100.000 beregnet på to forskellige måder.
Oversigt over DNEL baseret på en tærskelbaseret virkningsmekanisme og eksponerings-
niveauer, der resulterer i ekstra kræftrisikoniveauer på 1: 1000, 1:10 000 og 1: 100 000 baseret på
en ikke-tærskelbaseret virkningsmekanisme.
Virkningsmekanisme
Tærskel-baseret
Ikke tærskel-baseret
DNEL
Ekstra
kræftrisiko
1:1 000
1:10 000
1:100 000
Forslag til grænseværdi for TiO
2
NM
Inflammation
Lungekræft
Lungekræft
(metode I)
(metode II)
10 µg/m
3
4 µg/m
3
0.4 µg/m
3
0.04 µg/m
3
47 µg/m
3
4.7 µg/m
3
0.47 µg/m
3
Begge undersøgelser, som blev anvendt til risikovurderingen, benyttede P25 TiO
2
NM
(15-40 nm diameter, 80% anatase / 20% rutil). TiO
2
NM'er er forskellige med hensyn til
størrelse og overflade, men også coating, form, krystalstruktur mv. Den nærværende
arbejdsgruppe bemærker, at der er begrænsede tilgængelige data om de biologiske
effekter af forskellige fysisk-kemiske egenskaber, men arbejdsgruppen konkluderer, at
størstedelen af de tilgængelige data støtter, at overfladearealet (og derfor også størrelsen)
af TiO
2
er en prædiktor for partikelinduceret inflammation og akutfaserespons i
lungerne. NIOSH har vist, at partikeloverfladearealet af TiO
2
-partikler af forskellige
størrelser (fin og ultrafin) og forskellige krystalstrukturer (80% anatase/20% rutil og 99%
rutil) kan forklare den observerede variation i TiO
2
-partikelinduceret lungeinflammation
og lungekræft i rotteinhalationsundersøgelser. Dette understreger vigtigheden af
overfladearealet som en prædiktor for det inflammatoriske respons og
kræftfremkaldende effekt.
Den nærværende arbejdsgruppe betragter kræft som den vigtigste effekt.
DNEL-tilgangen er stærkt afhængig af antagelsen om en tærskeleffekt for inflammation
og kræft. Den nuværende arbejdsgruppe er af den opfattelse, at der stadig er usikkerhed
om, hvorvidt dette er tilfældet for TiO
2
NM induceret kræft.
Der blev anvendt to forskellige metoder til beregning af den overskydende risiko for
lungekræft baseret på den samme kroniske inhalationsundersøgelse. Ved den første
viii
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
metode blev den lungedeponerede dosis brugt til at estimere eksponeringsniveauerne.
Ved den anden metode blev luftkoncentrationerne anvendt direkte. Uafhængigt af den
anvendte metode til risikovurderingen, er de beregnede forslag til grænseværdier alle
meget lave. Disse niveauer er mere end 100 gange lavere end den nuværende danske
grænseværdi i arbejdsmiljøet for titanium på 6 mg/m
3
(målt som Ti svarende til 10
mg/m
3
TiO
2
).
Den nærværende arbejdsgruppe anbefaler metoden, hvor den overskydende risiko for
lungekræft baseres på lungedeponeret dosis, da denne tilgang tager højde for den
faktiske lungedeponering. Således er den forventede overskydende lungekræftrisiko i
forbindelse med erhvervsmæssig udsættelse for TiO
2
NM 1: 1 000 ved 4 µg/m
3
, 1:10 000
ved 0,4 µg/m
3
og 1: 100 000 ved 0,04 µg/m
3
TiO
2
NM.
ix
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
C
ONTENTS
Foreword ....................................................................................................................................... iii
Executive summary ...................................................................................................................... iv
Contents .......................................................................................................................................... x
Abbreviations ............................................................................................................................... 11
Introduction.................................................................................................................................. 13
Human studies............................................................................................................................. 15
Human exposure ..................................................................................................................... 15
Epidemiological studies ......................................................................................................... 15
Toxicokinetics .............................................................................................................................. 20
Animal studies ............................................................................................................................. 22
Rodent versus human response ............................................................................................ 22
Intratracheal instillation versus inhalation .......................................................................... 22
Selection of studies and endpoints ....................................................................................... 23
Pulmonary inflammation ....................................................................................................... 23
Genotoxicity and cancer ......................................................................................................... 25
Cardiovascular effects............................................................................................................. 28
Reproductive toxicity .............................................................................................................. 29
Mechanisms of toxicity ............................................................................................................... 31
Pulmonary inflammation, genotoxicity and cancer ........................................................... 31
Cardiovascular effects............................................................................................................. 32
Dose-response relationships .................................................................................................. 33
Particle characteristics............................................................................................................. 34
Previous risk assessments of TiO
2
............................................................................................. 35
IARC .......................................................................................................................................... 35
ENRHES.................................................................................................................................... 35
NEDO ........................................................................................................................................ 36
NIOSH....................................................................................................................................... 36
Scaffold project ........................................................................................................................ 37
ECHA’s Committee for Risk Assessment (RAC) ................................................................ 37
Scientific basis for setting an occupational exposure limit .................................................... 40
Endpoint: Inflammation ..................................................................................................... 40
Endpoint: Cancer ................................................................................................................. 41
Conclusion .................................................................................................................................... 45
x
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
A
BBREVIATIONS
3-ClTyr
3-NOTyr
5-OHMeu
8-OHdG
8-OHG
AF
Apo-A1
ApoE
BAL
BMD
BMDL
C3
CI
CNT
CRP
DNEL
ECHA
EBC
eNOS
HDL
Hs-CRP
IARC
ICAM-1
ICP-MS
IL
INEL
IP
LDL
LOAEC
MAD
MMAD
MWCNT
NIOSH
NM
NO
NOAEC
NFA
OEL
o-Tyr
RAC
REACH
REL
ROS
RR
SAA
3-chlorotyrosine
3-nitrotyrosine
5-hydroxymethyl uracil
8-hydroxy-2-deoxyguanosine
8-hydroxyguanosine
Assessment factor
Apolipoprotein A1
Apolipoprotein E
Broncho alveolar lavage
Benchmark dose
Benchmark dose lower bound
Complement factor 3
Confidence interval
Carbon nanotube
C reactive protein
Derived-no-Effect Level
European Chemicals Agency
Exhaled breath condensate
Endothelial nitric oxide synthase
High density lipoprotein
High-sensitivity C reactive protein
The International Agency for Research on Cancer
Intercellular cell adhesion molecule-1
Inductively Coupled Plasma Mass Spectrometry
Interleukine
Human indicative no-effect levels
Intraperitoneal
Low-density lipoproteins
Lowest observed adverse effect concentration
Malondialdehyde
Mass median aerodynamic diameter
Multi-walled carbon nanotube
National Institute for Occupational Safety and Health
Nanomaterial
Nitrogen oxide
No observed adverse effect concentration
National Research Centre for the Working Environment
Occupational exposure limit
o-tyrosine
ECHA’s Committee for Risk Assessment
Registration, Evaluation, Authorization and Restriction of Chemicals
Recommended exposure limit
Reactive oxygen species
Relative risk
Serum amyloid A
11
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
SCCS
SMR
SOD
SP-D
TiO
2
TNF
TWA
VCAM-1
Scientific Committee on Consumer Safety
Standardized mortality ratio
Superoxide dismutase
Surfactant protein D
Titanium dioxide
Tumour necrosis factor
Time-weighted average
Vascular cell adhesion molecule-1
12
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
I
NTRODUCTION
Titanium dioxide (TiO
2
) is a white solid inorganic and poorly soluble compound. TiO
2
in
various particle sizes including nanosizes have been used for almost 100 years in a
diverse range of industrial and consumer products. TiO
2
is used as white pigment in e.g
paints and as food colorant. Traditionally, TiO
2
has been considered a low toxicity
particles (Oberdorster et al. 2005). For that reason TiO
2
has previously been used as a
negative control particle in many animal studies. However, this view was changed with
studies showing lung cancer in rats following chronic exposure to a high dose of fine
TiO
2
(Lee et al. 1985) and a lower dose of TiO
2
nanomaterial (TiO
2
NM)(Heinrich et al.
1995). At the same time, toxicological studies showed that smaller TiO
2
particles induced
more inflammation than larger TiO
2
particles (reviewed by (Stone et al. 2017)). This
observation was followed up by toxicological studies of other types of low solubility
particles showing that more inflammation was induced by smaller particles compared to
larger particles with the same chemistry (Oberdorster et al. 2005).
The EU has adopted the following definition of a NM “A natural, incidental or
manufactured material containing particles, in an unbound state or as an aggregate or as
an agglomerate and where, for 50 % or more of the particles in the number size
distribution, one or more external dimensions is in the size range 1 nm - 100
nm.”(European Commission 2017). TiO
2
NMs differ regarding size and surface area but
also coating, shape, crystal structure etc. (OECD Environment 2016;NIOSH 2011).
In 2006, the International Agency for Research on Carcinogenicity (IARC) classified TiO
2
as possibly carcinogenic to humans (group 2B). This classification was based on
sufficient evidence of carcinogenicity in experimental animals and insufficient evidence
in humans. IARC does not differentiate between nano- and fine particles in their
classification (IARC 2010).
In 2011, the National Institute for Occupational Safety and Health (NIOSH)
recommended that exposure limits for TiO
2
are set based on their size: NIOSH
recommends exposure limits (RELs) of 2.4 mg/m
3
for fine TiO
2
and 0.3 mg/m
3
for
ultrafine TiO
2
. These RELs are estimated to equal lung cancer risk below 1 in 1,000
during working lifetime. Due to the lack of epidemiological studies of TiO
2
, NIOSH
choses to base the RELs on chronic rat inhalation studies and extrapolation to human
risk (NIOSH 2011).
To our knowledge, there are no legally binding NM-specific occupational exposure
limits (OELs) for TiO
2
. The present Danish OEL for titanium is 6 mg/m
3
(as Ti,
corresponding to 10 mg/m
3
for TiO
2
), and is regulated by the Danish Working
Environment.
The aim of the present report is to investigate if the present knowledge allows for a
suggestion of a health-based nanospecific OEL for TiO
2
NM
.
In this document we review
the relevant literature on the adverse effects of TiO
2
NM. The risk assessment
methodology of this report will follow the guidelines suggested by REACH (ECHA
2012). First, threshold or non-threshold effects are determined. Threshold effect assumes
13
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
that the organism can withstand a certain dose before adverse effects occur, whereas
non-threshold effects assume that any exposure to the substance can result in adverse
effects. For an OEL based on threshold effects, the following traditional approach is
utilized: 1) identification of critical effect, 2) identification of the NOAEC, 3) calculation
of OEL using assessment factors adjusting for inter and intra species differences. For
non-threshold effects, the present working group will use two different approaches for
calculating excess lung cancer risk. In the first approach lung burden will be used to
estimate the exposure levels. In the second approach, air concentrations were used
directly. Conclusively, the calculated OELs will be compared and lastly, a recommended
OEL for TiO
2
NM exposure will be proposed.
14
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
H
UMAN STUDIES
Human exposure
There are limited data on occupational exposure TiO
2
NM. A review of reported
exposure to different types of engineered NMs included 29 exposure scenarios of TiO
2
NM exposure. TiO
2
NM exposure occurred in research laboratories, industrial-scale
synthesis units, in a pilot-scale synthesis unit, in a laboratory-scale production unit, and
in a university research laboratory. The mass concentrations of respirable TiO
2
in the
workers’ breathing zone ranged from 10 to 150 µg/m
3
during bag filling. TiO
2
NM was
mainly detected as aggregated structures in a size range spanning from nanometer to
micrometer (Debia et al. 2016).
A recent exposure assessment, which was not included in the review by Debia et al., was
performed at a Chinese TiO
2
manufacturing plant. Mass concentrations were assessed in
two different worksites at the plant: In the packaging workshop, total dust
concentrations were 3.17 mg/m
3
of which 1.22 mg/m
3
was dust in the nanosize range. In
the milling workshop, total dust concentrations were 0.79 mg/m
3
of which 0.31 mg/m
3
was dust in nanosize range. ICP-MS analysis showed that a rather small part of the dust
was TiO
2
: TiO
2
content in total dust was 46.4 µg/m
3
at the packaging workshop and 39.4
µg/m
3
at the milling workshop. TiO
2
NM constituted 16.7 and 19.4 µg/m
3
, respectively
(Xu et al. 2016).
Epidemiological studies
A few epidemiological studies have been performed to evaluate the adverse effects of
inhalation exposure to TiO
2
in humans. None of the epidemiological studies included
information on the size range of the TiO
2
particles. Thus, it is not possible to determine
whether the exposures included TiO
2
NM. In total eight studies were identified of which
five are cohort studies (Chen and Fayerweather 1988;Fryzek et al. 2003;Boffetta et al.
2004;Ellis et al. 2010;Ellis et al. 2013) and three are case-control studies (Siemiatycki, 1991
(as referred by (IARC 2010) 2010 and (NIOSH 2011); (Boffetta et al. 2001),(Ramanakumar
et al. 2008).
Except for the newest studies by Ellis et al. (Ellis et al. 2010;Ellis et al. 2013), the studies
are included in the evaluations of TiO
2
performed by IARC (IARC 2010) and/or NIOSH
(NIOSH 2011). The present working group refers to these publications for a more
detailed description of these studies. The evaluation of TiO
2
performed in 2006 by IARC
concluded that there was inadequate evidence of carcinogenicity in humans (IARC
2010). The evaluation of TiO
2
by NIOSH in 2011 concludes that “these studies provide no
clear evidence of elevated risks of lung cancer mortality or morbidity among those
workers exposed to TiO
2
dust”(NIOSH 2011).
Among the studies included in NIOSH and/or IARC, Chen and Fayerweather (Chen and
Fayerweather 1988), Fryzek et al. (Fryzek et al. 2003) and Boffetta et al. (Boffetta et al.
2004) in addition to all cause and lung cancer also assessed death caused by
cardiovascular diseases.
15
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
The number of deaths from ischemic heart disease and cerebrovascular disease was
numerically increased among employees with TiO
2
exposure (n = 1 576) as compared
with employees without TiO
2
exposure (n=901) at two TiO
2
producing plants (Dupont,
US). However, this increase was not statistically significant at p<0.10. When the TiO
2
exposed workers were compared with the US reference group, the number of deaths
from diseases of the circulatory system was slightly decreased (Chen and Fayerweather
1988).
In the cohort study by Fryzek et al. (Fryzek et al. 2003) of 4 241 TiO
2
exposed workers at
four US TiO
2
plants, no significantly increased standardized mortality ratio (SMR) was
found for heart disease or cerebrovascular disease or for any other specific cause of death
as compared to the general background population.
In the European cohort study by Boffetta et al. (Boffetta et al. 2004) of 15 017 workers
employed at factories producing TiO
2
, no statistically significant increase in number of
deaths from cerebrovascular disease was found as compared with the general
background population.
The study by Ellis et al. (Ellis et al. 2010) investigated the mortality among workers
employed for at least 6 months in three DuPont TiO
2
plants in the United States (n=5054).
The general US population was used as reference. The mortality from all causes, lung
and larynx cancer, non-malignant respiratory disease and all heart disease was
statistically significantly decreased compared to the general population. The number of
cancers belonging to the category “Other respiratory cancers” was increased 2.5 fold
(95% CI: 0.62-6.46) compared to the general population. However, this was based on
only 3 cases and was not statistically significant. The very low standardized mortality
rates are characteristic when occupational active persons are compared with the general
population due to the healthy worker effect.
A second study by Ellis et al. (Ellis et al. 2013), sponsored by E.I. du Pont de Nemours
and Company, investigated the mortality among 3607 workers employed in the same
three DuPont TiO
2
plants as in the previous study by Ellis et al. (Ellis et al. 2010). The
study cohort overlapped with the 5054 workers in the previous study. Compared to the
previous study, stricter inclusion criteria were applied: In addition to having been
employed for at least 6 months, the job held had to have potential TiO
2
exposure, and no
more than 25% or 5 years missing job history was accepted. The outcomes were death
from all causes, all cancer, lung cancer, non-malignant respiratory disease, and all heart
disease. The number of employees and the age of the study group differed vastly
between the three factories which were included in the study. The employees at the
Edgemoor plant contributed with 56% of the person-years for follow-up, and 85% of the
deceased. Employees at Edgemoor plant were on average born in 1935, whereas
employees were on average born in 1952 and 1958, respectively, on the two other plants.
Consequently, the observed associations were driven by the Edgemoor plant, which
contributed with the largest study group and the most cases. The study used two
different reference groups, the US population and a control group of other Dupont
workers who were not exposed to TiO
2
. The present working group is of the opinion that
the reference group of non-exposed Dupont workers is the most appropriate reference
16
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
group, since it is most comparable to the TiO
2
exposed Dupont workers regarding health
worker effects, lifestyle factors and level and type of medical insurance, as compared to
the general population in the US. However, the working group notes that the Dupont
workers in the reference group may be exposed to other hazardous agents but no
information regarding this is provided in the publication. There were no statistically
significant differences in mortality for any of the studied outcomes for the TiO
2
exposed
workers compared to the US population. However, when the TiO
2
exposed workers
were compared to the reference group of other Dupont workers, an increased mortality
was observed for the following endpoints: all causes (SMR 1.23; 95% CI 1.15–1.32), all
malignant neoplasms (SMR 1.17; 95% CI 1.02–1.33), and lung cancer (SMR 1.35; 95% CI
1.07–1.33). The associations were driven by an increased mortality found at the
Edgemoor plant. Increased heart mortality was seen on the Edgemoor plant when this
plant was compared to “other workers” as reference group, but not for all three plants
combined. The risk estimates did not show clear dose-response relationship with
increasing cumulative dose. Risk estimates for all causes and non-malignant respiratory
disease increased marginally with increasing cumulative exposure using a 10 year lag,
and risk estimates for all cancers and non-malignant respiratory disease increased
marginally with increasing cumulative exposure using no lag. The observed difference in
the SMRs using an employed population versus a general population as a reference
group is typically associated with healthy worker effect. The studies by Ellis et al. (Ellis
et al. 2010;Ellis et al. 2013) have some severe study limitations including lack of
information on TiO
2
particle size, smoking history and a lack of a description of how the
reference groups were selected (including the number of persons in the reference
groups). Furthermore, there was no description of other possible occupational exposures
of the Dupont workers in the reference group.
While none of the above mentioned studies had information on particle size, the adverse
effects of TiO
2
in the nanosize have been specifically studied in two human
biomonitoring studies:
In one study, biomarkers of inflammation, oxidative damage of nucleic acids, proteins
and lipids were analyzed in the exhaled breath condensate (EBC) of a cohort of TiO
2
NM
manufacturing workers (n=36). The controls were healthcare personnel and technical
staff who were not employed at the factory and did not handle dusts (n=45). In the TiO
2
workshops, the median TiO
2
mass concentrations varied between 0.40 and 0.60 mg/m
3
.
In the facility, the median particle number concentration was approximately 2 x 10
4
particles/cm
3
of which approximately 80% of the particles were less than 100 nm. In the
research workspace, the air concentration (0.16 mg/m
3
) and the particle number (1.32 *
10
4
particles/cm
3
) were lower. The results have been published in a series of articles and
documented inflammation (Pelclova et al. 2016b), oxidative damage of nucleic acids and
proteins (Pelclova et al. 2016a) and lipid oxidative damage (Pelclova et al. 2017) in the
EBC of the cohort of TiO
2
NM manufacturing workers. TiO
2
concentrations in the EBC
were statistically significantly increased in the production workers (~20 µg TiO
2
/L,
p<0.001)) compared to both research personnel (2.00 µg/L) and controls (1.12 µg/L). The
levels of oxidative stress markers (8-OHdG, 8-OHG, 5-OHMeu, o-Tyr, 3-ClTyr, 3-
chlorotyrosine and 3-NOTyr were higher in the production workers than the workers
from the research wing of the plant and unexposed controls (Pelclova et al. 2016a).
17
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
Inflammation was evaluated by measuring leukotriens in EBC. All of the measured
leukotrienes were statistically significantly increased compared to the control group
(Pelclova et al. 2016b). Lipid oxidation measured as malondialdehyde, 4-hydroxy-trans-
hexenal, 4-hydroxy-trans-noneal, 8-isoProstaglandin
F2α and aldehydes C
6
-C
12
was
increased in TiO
2
exposed workers compared to controls and a significant dose-response
relationship was found between exposure to TiO
2
and markers of lipid oxidation in the
EBC (Pelclova et al. 2017).
In another recent cross-sectional study, cardiopulmonary effects were analysed in
exposed workers (n=83) and controls (n=85) in a TiO
2
NM manufacturing plant in China.
The exposure is described in detail in (Xu et al. 2016) and is described in the above
paragraph on exposure in the present report. In short, the highest dust concentration was
measured in the packaging area where the total mass concentration of particles was 3.17
mg/m
3
of which 1.22 mg/m
3
was nanoparticles (39% of total mass). Only a minor part of
the dust was TiO
2
(46.4 µg/m
3
) and even less was TiO
2
NM (16.7 µg/m
3
). A number of
assessed markers of inflammation (IL-8, IL-6, IL-1β,
TNF-α, and IL-10),
oxidative stress
(SOD and MDA), cardiovascular disease (VCAM-1, ICAM-1, low-density lipoproteins
(LDL) and total cholesterol) and lung damage (surfactant protein D (SP-D) and
pulmonary function) were associated with occupational exposure to TiO
2
NM. The acute
phase proteins serum amyloid A (SAA) and high-sensitivity C reactive protein (hs-CRP)
and the inflammatory markers IL-1β
and IL-10
were also measured but for these markers
no significant differences between the groups were observed. Among the measured
biomarkers, SP-D was the only marker showing dose dependency: SP-D decreased with
increasing working time (Zhao et al. 2018).
IARC and NIOSH (IARC 2010;NIOSH 2011), concluded that the included
epidemiological studies did not show increased risks of lung cancer among workers
occupationally exposed to TiO
2
. However, in a recent study of workers exposed to TiO
2
at three different plants in the USA, statistically significantly elevated SMRs were found
for all causes, all cancers, and lung cancers when non-TiO2 exposed workers at other
Dupont factories were used as reference group while no increase was found when the
US population was used as reference group (Ellis et al. 2013). Mortality of heart disease
associated with TiO
2
exposure has been assessed by Chen et al. (Chen and Fayerweather
1988) and Ellis et al. (Ellis et al. 2010;Ellis et al. 2013). When using the US population or
other workers as reference group neither study showed increased heart mortality among
TiO
2
workers.
The present working group is of the opinion that the reference group of non-exposed
Dupont workers is the most appropriate reference group as compared to the general US
population, and therefore concludes that increased risk of all-cause mortality, malignant
neoplasms, and lung cancer was found in the study by Ellis et al. (Ellis et al. 2013).
In relation to assessing the effects of TiO
2
NMs, a major limitation is that none of the
studies on lung cancer and heart disease provided information on particle size.
However, the literature search identified two biomonitoring studies with information of
particle size. In these studies, biomarkers of effect were associated with occupational
18
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
2130137_0021.png
exposure to TiO
2
NM. The biomarkers reflect a local biological response to TiO
2
NM in
the pulmonary region of the exposed workers and a systemic response in the blood.
The ability to detect the effect of exposure to occupational carcinogens is also determined
by the population-specific lung cancer incidence. In Denmark, the life time risk of getting
lung cancer (0-74 years) is 4.9% for men and 4.5% for women, respectively, according to
The National Board of Health. In the US, life time lung cancer risk is similar, 7% for men
and 6% for women (American Cancer Society 2018). The relative lung risk caused by
occupational exposure to a carcinogen, which causes lung cancer the different risk levels,
1%, 0.1% and 0.01% are given in table 1. As can be seen in the table, exposures that cause
1% excess lung cancer will give relative risks of 1.2. According to power calculation,
detection of 1% excess cancer incidence with 5% lung cancer incidence in the reference
group would require group sizes of 8 000 participants (with 80% chance of detecting the
effect at 5% significance level). On the other hand, occupational exposures that cause
0.1% excess lung cancers (1 of 1 000, which is the acceptance level in the US),
corresponds to a RR of 1.04, which requires group sizes of 750 000 persons if the
background cancer incidence is 5%.
Table 1. Relative risk of lung cancer for carcinogens that cause 1%, 0.1% or 0.01% excess lung
cancer risk in a population with the current Danish lung cancer incidence
Life time risk (0-74 years)
2011-2015 in Denmark
1
Excess lung cancer risk
level
1:100
2:1000
1:1 000
1:10 000
1:100 000
Men
4.9%
RR
RR= (4.9+1)/ 4.9= 1.20
RR= (49+2)/49= 1.041
RR= (49+1)/49= 1.02
RR= (490+1)/490= 1.002
RR= (4900+1)/4900= 1.000 2
Women
4.5%
RR
RR= (4.5+1)/4.5= 1.22
RR= (45+2)/45=1.044
RR= (45+1)/45=1.02
RR= (450+1)/450= 1.002
RR= (4500+1)/4500= 1.000 2
Thus, the epidemiological studies on TiO
2
and lung cancer risk have limited statistical
power to detect carcinogenic effects of TiO
2
exposure, unless the excess lung cancer risk
associated with TiO
2
exposure was very high.
As none of the above mentioned epidemiological studies provided information on the
size range of the TiO
2
particles, thus making it impossible to determine whether the
exposures included TiO
2
NM, and no information on dose-response relationship, the
present working group has decided to include and base the suggested OEL of
experimental animal studies.
19
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
T
OXICOKINETICS
Exposure to TiO
2
NM may occur by one of three exposure routes: inhalation, ingestion,
or dermal. Of these, inhalation and to some degree dermal are the main exposure routes
in the occupational setting. Dermal exposure through healthy skin is most likely not a
risk following short term exposure. However, uptake may occur through damaged skin
or if exposure is chronic (Christensen et al. 2011). The focus of this report is exposure by
inhalation.
Inhalation of particles results in deposition in the respiratory tract (nasopharyngeal,
tracheobronchial and alveolar regions) (Oberdorster et al. 2005). The deposition pattern
of particles in the different parts of the respiratory tract is strongly dependent on size of
the aerosolized particle agglomerate. Inhaled NMs deposit in the entire respiratory tract.
However, a large fraction of the inhaled NMs deposit in the alveolar region. In contrast,
most of the larger particles (> 1-2
µm)
deposit in the upper airways (Oberdorster et al.
2005) (Shi et al. 2013;Koivisto et al. 2012). In a study, mice were exposed by inhalation
1h/day for 11 days to 42 mg/m
3
aerosolized powder of rutile TiO
2
with an average
crystallite size of 21 nm. The pulmonary deposition fraction was estimated to be 8.6%
based on the observed particle size distribution in the aerosol (Hougaard et al. 2010).
A 12 week inhalation study in rats showed that pulmonary clearance of 21 nm TiO
2
NM
was slower (t
1/2
= 501 days) than 250 nm TiO
2
particles (t
1/2
= 174 days) (Ferin et al. 1992). .
The main mechanism for particle clearance in the alveoli following TiO
2
NM inhalation
was phagocytosis by macrophages(Shi et al. 2013;Koivisto et al. 2012). Smaller particles
are less efficiently phagocytized than larger particles: A rat inhalation study with 20 nm
TiO
2
particles demonstrated that nanoTiO
2
particles are not efficiently phagocytized by
macrophages (Geiser et al. 2008). This results in prolonged residence time for particles in
the lungs increasing the possibility for inflammatory reactions and translocation into
lung tissue or the circulation. Ferin et al. showed that nanosized TiO
2
translocate from
the lungs to the blood circulation to a greater extent than larger TiO
2
particles (Ferin et al.
1992). As reviewed by Geiser & Kreyling, human studies have shown that the
translocated nanoparticle mass fraction is less than 1 % of the dose delivered to the lungs
(Geiser and Kreyling 2010).
Only few studies have measured translocation of TiO
2
NM from the lung into the
circulatory system to systemic tissue. Available data suggest that the rate of NM
migration to the circulatory system is low. The rate of translocation is likely to depend
on size, shape and surface modifications (Geiser and Kreyling 2010).
In a very comprehensive study, Kreyling and co-workers studied the biokinetics of
radiolabeled 70 nm TiO
2
NM following intratracheal instillation (Kreyling et al. 2017b),
intravenous injection (Kreyling et al. 2017a) and oral application (Kreyling et al. 2017c) in
rats. For the intratracheal and the intravenous studies, biodistribution was assessed
quantitatively 1 h, 4 h, 24 h, 7 d and 28 d after exposure. The biodistribution following
oral application was assessed on the same time points except for the latest time point.
20
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
The intratracheal instillation study showed that after 1 h about 4% of the initial
peripheral lung dose had translocated from the pulmonary region. The TiO
2
NM were
mainly retained in the carcass (4% after 1 h and 0.3% after 28 d). In the liver and kidney
the fractions of TiO
2
NM remained constant (0.03%) (Kreyling et al. 2017b). A
comparison of the biodistribution after IV-injection (Kreyling et al. 2017a), gavage
(Kreyling et al. 2017c) and intratracheal instillation (Kreyling et al. 2017b), showed that
gavage and intratracheal instillation resulted in a similar patterns of biodistribution.
However, the rate translocation to secondary organs was higher following pulmonary
exposure (ca. 4.3% of the pulmonary deposited dose after 1 h) compared to oral exposure
(0.6% of the administered dose passed the gastro-intestinal-barrier after one hour). The
biodistribution following intravenous injection was very different from the
biodistribution of following pulmonary and oral dosing.
21
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
A
NIMAL STUDIES
Rodent versus human response
Inhalation studies in mice and rats are used to assess potential human hazard where
human exposure studies and epidemiological studies are not available.
There is very limited data available on effects following inhalation of TiO
2
NMs in
humans. Rats are the preferred animal model in particle toxicology and are more
sensitive than mice to particle-induced lung cancer and fibrosis.
Intratracheal instillation versus inhalation
Inhalation studies are the gold standard of toxicity testing, as this exposure route is the
closest surrogate to human exposure. However, the deposited pulmonary dose can be
difficult to monitor after inhalation due to differences in sizes of the aerosolized particle
agglomerates. This can result in differences in deposition (Schmid and Cassee 2017). In
addition, exposure by inhalation requires a substantial amount of material and
specialized inhalation facilities, and it poses an occupational health risk to the
technicians handling the NMs.
Pulmonary deposition by intratracheal instillation is used in screening studies (Bourdon
et al. 2012;Husain et al. 2013;Poulsen et al. 2015b;Saber et al. 2012b;Saber et al. 2012a) and
has been proposed as an alternative to inhalation exposure. This exposure method
ensures that the same dose is delivered to the lung for all NM exposures, demands less
material and is more user-friendly. Intratracheal installation has previously been shown
to give widespread distribution of particles throughout the lung (Mikkelsen et al. 2011).
A number of studies have compared the toxicological response following inhalation and
instillation of nanomaterials. Two studies have compared the global transcriptional
profiles as a means to investigate the pulmonary biological response after inhalation
compared to instilled or aspirated NMs. Inhalation and intratracheal instillation of a
surface modified TiO
2
NM resulted in similar transcriptional changes, with the acute
phase response and inflammation as the most important pulmonary responses to inhaled
and instilled TiO
2
(Halappanavar et al. 2011;Husain et al. 2013). Similarly, Kinaret et al
(Kinaret et al. 2017) compared the global transcriptomic profiles of lung tissue from mice
exposed to a straight and long multi-walled carbon nanotube (MWCNT) by inhalation or
aspiration. The authors concluded that the perturbed pathways were very overlapping,
suggesting that the transcriptiomic response to MWCNT exposure was very similar for
inhaled and pulmonary dosed MWCNTs.
Other studies compared levels of pulmonary inflammation, measured as neutrophil
influx, after exposure by inhalation or intratracheal instillation in rodents. Two studies
using MWCNT reported that both methods resulted in pulmonary inflammation, with
inhalation being more potent at inducing inflammation (Morimoto et al. 2012;Porter et al.
2013). Baisch et al. reported that instillation of a high dose of TiO
2
nanoparticles induced
greater inflammation compared to low dose rate delivery through inhalation, even
though the same pulmonary deposited dose were delivered. The authors concluded that
22
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
intratracheal instillation is useful for quantitative ranking of nanoparticle hazards, but
not for quantitative risk assessment (Baisch et al. 2014).
Selection of studies and endpoints
In the present report inhalation studies will be prioritized. For the description of
toxicological endpoints and mechanism of toxicity, studies using pulmonary deposition
as intratracheal instillation will be included where no quality inhalation studies are
available. Dose-response assessments, however, are solely conducted based on sub-
chronic and chronic inhalation studies.
Endpoints were evaluated based on reported adverse effects of TiO
2
NM exposure in
reports and in the scientific literature. The assessment by NIOSH used cancer as the
endpoint (NIOSH 2011). However, other previous assessments have mainly focused on
inflammation as critical effect (Christensen et al. 2011;Stockmann-Juvela et al.
2014;Nakanishi and Gamo 2011). This report will therefore include both endpoints.
Cancer and cardiovascular disease have been identified as two of the main mortality
causing diseases in the world (World Health Organization 2018;Cancer Risks UK 2018).
Both diseases are potentially initiated by inflammation, as described in
Mechanism of
toxicity.
In conclusion, the critical endpoints were chosen based on literature review and
mechanistic understanding.
Pulmonary inflammation
In a sub-chronic inhalation study by Ferin et al., rats were exposed to about 23 mg /m
3
of
two different sized anatase TiO
2
particles (21 nm and 250 nm) for 6 h/day, 5 days/week
for 12 week. Pulmonary inflammation was assessed 4, 8, 12, 41 and 64 weeks after start
of exposure. The 21 nm TiO
2
NM induced more neutrophil influx than the 250 nm TiO
2
particles and the filtered air already after 4 weeks of exposure. The number of
neutrophils were almost reduced to control level after 52 weeks post-exposure (Ferin et
al. 1992).
In a sub-chronic inhalation study by Bermudez et al, female rats, mice and hamsters in
groups of 25 were exposed to 0, 0.5, 2.0 or 10 mg/m
3
TiO
2
NM(P25, average primary
particle size of 21 nm) for 6 hours/day, 5 days/week for 13 weeks (Bermudez et al. 2004).
The mean mass-median aerodynamic diameter of TiO
2
NM and agglomerates was 1.37
µm
in exposure chamber. Pulmonary endpoints (inflammation, cytotoxicity, lung cell
proliferation and histopathology) were assessed 0, 4, 13, 26 and 52 weeks (49 weeks for
TiO
2
NM exposed hamsters) after end of exposure.
To assess inflammation, the total number of broncho alveolar lavage (BAL) cells and the
number of macrophages, neutrophils, eosinophils and lymphocytes in the BAL cells was
determined. The neutrophil influx as percent of total BAL cells is shown in Table 2.
Compared to controls, the percentage of neutrophils was not significantly increased in
hamsters at any dose or time point after end of exposure. Immediately after end of
exposure rats had increased percentage of neutrophils at 2 mg/m
3
and above while this
23
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
was not the case for exposure at 0.5 mg/m
3
. From 4 weeks after exposure and later time
points, both rats and mice had increased percentage of neutrophils at the highest dose
(10 mg/m
3
).
In rats exposed at the highest dose (10 mg/m
3
), progressive epithelial and
fibroproliferative changes were observed through 13 weeks post-exposure. Most of these
changes were reported to be regressing over time (13-52 weeks post-exposure).
Inhalation and intratracheal instillation studies have shown that when rats and mice
were exposed TiO
2
, the ultrafine TiO
2
induced a much stronger pulmonary inflammatory
response compared to the same mass of fine TiO
2
particles. The inflammatory response
correlated with the surface area of the deposited particles irrespectively of size. This dose
response relationship has been observed for a number of low-toxicity low – solubility
particles and it is generally accepted that the inflammatory response of low toxicity-low
solubility particles including TiO
2
is proportional to the surface area of the instilled
particles rather than the mass (reviewed in Oberdörster et al (Oberdorster et al. 2005)).
We consider the Bermudez study (Bermudez et al. 2004) as a key study for this hazard
assessment. It is the only of the identified studies that is a sub-chronic inhalation study
with dose-response relationship of TiO
2
NM. In that study, a NOAEC of 0.5 mg/m
3
was
identified for pulmonary influx of neutrophils in rats which was the most sensitive of the
tested species. In addition to this study we have identified a range of inhalation studies
with shorter exposure duration using TiO
2
NM particles (Ma-Hock et al. 2009;Noel et al.
2012;Rossi et al. 2010a;Rossi et al. 2010b;Baisch et al. 2014;Kwon et al. 2012;Lindberg et al.
2012). Overall they support that a NOAEC level for pulmonary influx of neutrophils is in
the range of 0.5-2 mg/m
3
.
24
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
2130137_0027.png
Table 2. Pulmonary influx (%) of neutrophils in animals exposed by inhalation to TiO
2
NM
(Bermudez et al. 2004).
Postexposure Concentration
(weeks)
0
(mg/m
3
)
0
0,5
2
10
0
0,5
2
10
0
0,5
2
10
0
0,5
2
10
0
0,5
2
10
Rats
0.4 ±0.42
0.5 ± 0.35
6.50 ± 4.23*
64.80 ±5.35*
0.3 ± 0.27
0.2 ± 0.27
0.90 ± 0.96
43.30 ± 3.27*
0.20 ± 0.27
1.20 ± 1.15
1.70 ± 1.60
41.9 ±14.10*
0.50 ±0.35
0.30 ± 0.27
1.90 ± 0.82
20.8 ± 7.64*
0.80 ± 0.84
0.60 ± 0.42
0.70 ± 0.57
12.00 ± 4.51*
Mice
0.00 ± 0.00
0.00 ± 0.00
0.20 ± 0.27
14.50 ± 5.73*
0.20 ± 0.27
0.20 ± 0.27
0.10 ± 0.22
12.40 ± 7.90*
0.10 ± 0.22
0.40 ± 0.55
0.30 ± 0.45
13.90 ± 6.81*
0.10 ± 0.22
0.30 ± 0.27
0.10 ± 0.22
17.00 ± 5.95*
Hamsters
2.30 ± 1.15
1.30 ± 6.84
1.00 ± 0.79
10.20 ± 14.65
2.30 ± 2.49
5.20 ± 4.72
4.10 ± 4.60
3.70 ± 3.49
7.80 ±
2.60 ±
2.60 ±
2.70 ±
4.70 ±
3.10 ±
3.60 ±
2.30 ±
8.24
1.71
0.89
1.25
1.68
2.70
1.47
1.96
4
13
26
0.10 ± 0.22
4.50 ± 2.50
0.00 ± 0.00
5.50 ± 6.28
0.40 ± 0.65
5.20 ± 3.19
12.30 ± 4.80*
4.50 ± 1.70
*Significantly different from control, p < 0.05; NOAECs and
the lowest observed adverse
effect concentrations (
LOAECs
)
for each species at different time-points after end of
exposure are indicated with green and red text, respectively. Table is generated based on
Tables S1-S3 in Bermudez et al (Bermudez et al. 2004).
52
Genotoxicity and cancer
Genotoxicity and cancer are well studied, possible adverse effects of exposure to TiO
2
NM. Genotoxicity often occurs relative rapidly after exposure, whereas cancer is a more
complex pathological endpoint that requires longer time to develop. In this report, we
therefore chose to differentiate between genotoxicity in shorter-term studies and cancer
in long-term studies.
Cancer
Three chronic cancer TiO
2
inhalation studies were identified. One of these studies used
TiO
2
NM (P25, 80%anatase/20% rutile) which was tested in female Wistar rats (Heinrich
et al. 1995;Heinrich et al. 1995), while the other two studies used both fine, rutile TiO
2
but tested in male/female Wistar rats (Muhle et al. 1991) and in male/female Sprague-
25
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
Dawley rats (Lee et al. 1985), respectively. Details on the study set-ups are summarized
in Table 3. An increased cancer incidence was detected in rats exposed to 10 mg/m
3
of
TiO
2
NM (Heinrich et al. 1995), while exposure to fine TiO
2
only increased cancer
incidence in rats exposed to the highest tested concentration (250 mg/m
3
) (Lee et al.
1985). In rats exposed to 10 mg/m
3
of TiO
2
NM, slight to moderate interstitial fibrosis in
the lungs was observed in all animals after 2 years of exposure (Heinrich et al. 1995). The
present working group notes that dose response relationship for TiO2 NM-induced
cancer could not be established since only one dose level was tested. However, NIOSH
has evaluated the rat cancer data from inhalation studies of TiO
2
in different sizes
(ultrafine and fine) and concluded that they fit on the same dose-response curve when
dose is expressed as total particle surface area in the lungs (NIOSH 2011). The present
working group considers this sufficient evidence of dose response relationship.
In summary, in the only identified chronic inhalation study of rats exposed to TiO
2
NM,
cancer was induced at 10 mg/m
3
(LOAEC = 10 mg/m
3
). TiO
2
has been classified as
possibly carcinogenic by IARC based on sufficient evidence of carcinogenicity in
experimental animals (IARC 2010). When NIOSH evaluated rat cancer data from
inhalation studies of TiO
2
in different sizes (ultrafine and fine) they concluded that all
the data points fit on the same dose-response curve when dose was expressed as total
particle surface area in the lungs (NIOSH 2011).
26
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
2130137_0029.png
Table 3. Overview of chronic rat inhalation studies
Reference
Muhle et al.
(Muhle et al.
1991)
Lee et al. (Lee
et al. 1985)
(reclassification
of tumors in
Warheit and
Frame (Warheit
and Frame
2006))
Type of TiO
2
Fine, rutile
Exposure
2 year, 0, or 5
mg/m
3
Whole body
inhalation for 6
hour/day, 5
days/week for
up to 2 years,
to 0, 10, 50, or
250 mg/m
3
No follow up
time after end
of exposure.
Lung tumor increase compared to controls
5 mg/m
3
: No increase
Fine, rutile
10 mg/m
3
: No increase
50 mg/m
3
: No increase
250 mg/m
3
: Increased
For the 250 mg/m
3
:
Bronchioalveolar carcinomas in 12/77 male
rats and 13/74 female rats
Squamous cell carcinomas in 1/77 males and
13/74 females)
Controls:
Bronchioalveolar carcinomas in 2/79 male
rats and 0/77 female rats
No squamous cell carcinomas.
10 mg/m
3
: Increased
At 30 months:
13/100: Adenocarcinoma
3/100: Squamous cell carcinoma
4/100: Adenocarcinoma
20/100: keratinzing cystic squamous-cell
tumors
32/100: Total number with tumors
Controls:
Adenocarcinomas: 1/217
No other lung tumors were observed
Heinrich et al.
(Heinrich et al.
1995)
Ultrafine P25
(15-40 nm
primary
particle size, 0.8
µm MMAD, 48
m
2
/g specific
surface area,
80%
anatase/20%
rutile)
18 hour/day, 5
days/week for
up to 2 years to
0, or 10 mg/m
3
followed by 6
months
without TiO
2
exposure
Genotoxicity
The genotoxic potential has been tested in many
in vivo
studies by analysis of different
endpoints including DNA strand breaks, DNA adducts and micronuclei. Some studies
indicate that TiO
2
NMs are genotoxic, while other studies do not (reviewed by (Shi et al.
2013)). The different physico-chemical properties of the tested TiO
2
particles (specific
surface area, coating, anatase/rutile, form) may explain why some studies are negative
and others positive. The present working group concludes that no firm conclusions can
be reached regarding genotoxicity.
27
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
Cardiovascular effects
Only few studies have investigated the cardiovascular effects of pulmonary TiO
2
NM
exposure. Some studies have assessed promising biomarkers for cardiovascular disease.
Plaque progression and vascular dysfuction
The lipid profile of mice significantly differs from that of humans. Mice do not develop
atherosclerosis, because rapid clearance of hepatic LDL results in low and rather stabile
total serum cholesterol levels, even after increased cholesterol intake and synthesis.
Atherosclerotic changes are therefore mainly investigated in ApoE -/- mice, which are
deficient in apolipoprotein E (apoE), a glycoprotein associated with all lipoproteins
except LDL. ApoE -/- mice develop spontaneous atherosclerosis as early as 3–4 months
of age when fed normal chow (Nakashima et al. 1994). This makes them suitable for
investigating cardiovascular effects.
A few studies have reported TiO
2
NM-induced accelerated plaque progression:
Modest effect on plaque progression was detected in ApoE-/- mice intratracheally
instilled with 0.5 mg/kg bodyweight TiO
2
NM (21 nm) once a week for 4 weeks. No
effect on vasodilatory function was detected in ApoE-/- mice intratracheally instilled
with 0.5 mg/kg bodyweight of three types of TiO
2
(rutile 288 nm TiO
2
, anatase/rutile 12
nm TiO
2
, and rutile 21 nm TiO
2
) at 26 and 2 hours before measurement (Mikkelsen et al.
2011).
ApoE-/- mice were exposed by tracheal instillation of 0, 10, 50 and 100 µg 5-10 nm TiO
2
NM once a week for 6 weeks. Compared to vehicle controls, the high dose group had
increased levels of CRP, nitrogen oxide (NO), endothelial nitric oxide synthase (eNOS),
total and high density lipoprotein (HDL) cholesterol in serum. In addition, the medium
and high dose group had increased plaque area and increased ratio of the lipid-rich core
area to plaque area, respectively. At the highest dose, TiO
2
NM exposure induced
systemic inflammation (measured as increased level of Hs-CRP), endothelial dysfunction
(measured as reduced serum level of nitric oxide and eNOS) and changed lipid
metabolism (measured as increased total cholesterol and decreased HDL in the serum)
(Chen et al. 2013).
Microvascular dysfunction was observed in rats exposed by inhalation to TiO
2
NM. The
microvascular dysfunction was associated with increased oxidative stress and decreased
NO production (Nurkiewicz et al. 2009).
Thrombus formation
Systemic administration of a single dose (1 mg/kg) of anatase TiO
2
NM (38 nm, 320 m
2
/g)
but not rutile TiO
2
NM (67 nm, 60 m
2
/g) accelerated thrombus formation in the
microcirculation in mice (Haberl et al. 2015).
Activation of complement factor 3 (C3) may promote the atherosclerotic process because
C3 activation products (C3a and C3b) are involved in the atherothrombotic process and
they are associated with lipid components in the vessel wall. Activation of C3 in blood
28
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
was detected in C57BL/6 mice exposed by intratracheal instillation
to 18 or 162 µg of
TiO
2
NM (rutile, 21 nm) compared with vehicle controls (Husain et al. 2015).
Acute phase response
The acute phase response is induced in humans in response to infection, infarction and
trauma, and it is defined by increases in acute phase response proteins with the most
predominant being CRP, SAA, and fibrinogen. During an acute phase response these
proteins can increase thousand fold (Gabay and Kushner 1999). Elevated plasma levels
of CRP and SAA have been reported as a risk factor for cardiovascular disease in
humans (Johnson et al. 2004;Lowe 2001;Mezaki et al. 2003;Ridker et al. 2000). In mice, the
SAA isoforms are the main acute phase response proteins, while CRP is only moderately
induced by inflammatory stimuli (Whitehead et al. 1990;Pepys and Hirschfield 2003).
SAA (SAA1-4) is a highly conserved family of apolipoproteins associated with HDL.
Several studies have reported changes in
Saa
expression levels after pulmonary exposure
to TiO
2
NM. Inhalation of TiO
2
NMs as well as intratracheal instillation of a single dose
of TiO
2
NM (21 nm) in female C57BL/6 mice strongly increased
Saa1, Saa2
and
Saa3
mRNA levels in lung tissue in a dose-dependent manner (Saber et al. 2013;Halappanavar
et al. 2011;Husain et al. 2013). Time-mated mice were exposed by inhalation 1h/day to 42
mg/m
3
TiO
2
NM on gestation days 8–18.
Saa3
mRNA expression levels were increased 5
days and 4 weeks after the end of exposure (Saber et al. 2013).
Reproductive toxicity
Time mated female rats were exposed to TiO
2
NM (P25, 21 nm in diameter) by inhalation
for 8 non-consecutive days (4-6 h/day for 7.8 days). The mass concentration was 10
mg/m
3
. The
calculated cumulative, deposited dose was 217 ± 1.0
µg. Chromatin
immunoprecipitation and DNA sequencing were performed in the offspring fetal hearts
at gestation day 20; and it was reported that the experiments provide initial evidence
that significant epigenetic and transcriptomic changes occur in the cardiac tissue of
maternally TiO
2
NM exposed progeny (Stapleton et al. 2018a). Two other studies used a
similar dosing regimen. One found that gestational exposure to the 21 nm TiO
2
particles
(median aerodynamic diameter 130-150 nm) disrupted progeny cardiac function and
bioenergetics (Hathaway et al. 2017). In the other study, the median aerodynamic
diameter of the inhaled 21 TiO
2
NM particles was 171 nm and the calculated daily
maternal deposition was 13.9 ± 0.5 µg. At 5 months of age a standard battery of several
locomotion, learning, and anxiety tests was applied for testing of male offspring from
four control and four exposed dams (n=11). TiO
2
NM was associated with significant
working memory impairments in the radial arm maze and deficits in the visual platform
test, possibly reflecting deficits in initial motivation in male F1 adults (Engler-Chiurazzi
et al. 2016). In a final study from this research group, virgin and late stage pregnant [GD
19] female rats were exposed to TiO
2
NM (21 nm in diameter) by inhalation for 5 h. The
mass concentration was 10 mg/m
3
(median aerodynamic diameter particle diameter
173.2±6.4 nm in the exposure atmosphere), leading to a calculated deposited dose of 42.2
± 1.9 µg TiO
2
. Assessment in live animals showed that inhalation of TiO
2
NM disturbed
vascular reactivity differentially in different stages of estrous in non-pregnant females. In
addition, increased inflammatory activity was observed in these animals. The level of
29
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
inflammatory markers in blood was altered during estrus and late gestation. The authors
suggest that female fertility may be impaired by TiO
2
NM inhalation (Stapleton et al.
2018b).
Another research group exposed time-mated female mice to TiO
2
NM (UV Titan, 21 nm
in diameter) by inhalation for 1h/day on gestation days 8 to 18. The mass concentration
was 42 mg/m
3
and the major particle size-mode was ~100 nm. Several outcomes were
studied in the time-mated females and their offspring. TiO
2
NM exposure was associated
with lung inflammation in the time-mated females 5 and 27 days post-exposure. In the
adult offspring, TiO
2
NM exposure was associated with moderate neurobehavioral
alterations. Cognitive function was unaffected but the offspring tended to avoid the
central zone in the open field assay. In addition, exposed female offspring displayed
enhanced prepulse inhibition in the acoustic startle test (Hougaard et al. 2010). Levels of
DNA strand breaks were evaluated using the comet assay. No effects were observed on
this endpoint in BAL cells or liver of time-mated females 5 and 27 days post exposure,
nor in the livers of their offspring at postnatal day 2 and 22 (Jackson et al. 2013). At
maturity, female F1 offspring were mated with unexposed males. Expanded-simple-
tandem-repeat-loci germline mutation rates were determined in the F2-generation and
found not to differ between TiO
2
and control F2 offspring (Boisen et al. 2012). Also
testicles were collected from the mature F1 and F2 males. Daily sperm production was
not statistically significantly affected in the F1- or F2-generation males originating in
dam with TiO₂ exposure compared to sham exposed dams
(Kyjovska et al. 2013).
Overall, the above described developmental toxicity effects were observed after 8 days of
exposure for 4-6 h per day at 10 mg TiO
2
NM/m
3
, or following exposure to 42 mg TiO
2
NM/m
3
for 1 h per day for 11 days. The corresponding daily doses were 5-7.5 mg/m3 per
8-h workday. When taking into account the LOAEC/NOAEC observed on increased BAL
neutrophils in BAL in other studies at 2/0.5 mg/m
3
(E.g. (Bermudez et al. 2004)),
developmental toxicity is not deemed to be the critical effect in the current investigation.
This, however, may change if experiments with lower mass concentrations of TiO
2
are
undertaken in the future.
30
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
M
ECHANISMS OF TOXICITY
Pulmonary inflammation, genotoxicity and cancer
Pulmonary exposure to TiO
2
NM has consistently shown dose-dependent pulmonary
inflammation (NIOSH 2011)and deposited surface area has been identified as an
important predictor of pulmonary inflammation (NIOSH 2011). The present working
group notes that there is limited available data on the biological effects of TiO
2
NM with
different physico-chemical properties, but concludes that the majority of available data
support that the surface area (and therefore the size) of TiO
2
is a critical driver of
particle-induced inflammation in the lungs. The present working group concludes that
inhalation of TiO
2
NM induces dose dependent pulmonary inflammation and that
neutrophil influx is predicted by the total surface area of deposited particles. The
working group considers inflammation as a threshold effect.
Shi et al. reviewed TiO
2
NM-induced genotoxicity
in vivo
and
in vitro
and concluded
that:” The possible mechanisms for TiO
2
NM-induced genotoxicity involve DNA
damage directly or indirectly via oxidative stress and/or inflammatory responses”(Shi et
al. 2013).
IARC has classified TiO
2
as possibly carcinogenic to humans (group 2B) based on
sufficient evidence of carcinogenicity in experimental animals and insufficient evidence
in humans. IARC does not differentiate between nano- and fine particles in their
classification (IARC 2010).
NIOSH concludes that the inflammatory response and the induction of lung tumors by
TiO
2
and other low-toxicity low-solubility particles correlates well with the total surface
area of pulmonary deposited particles (NIOSH 2011). NIOSH furthermore concludes
that “TiO
2
is not a direct-acting carcinogen, but acts through a secondary mechanism that
is not specific to TiO
2
but primarily related to particle size and surface area (NIOSH
2011).
EU’s Scientific Committee on Consumer Safety (SCCS) concluded similarly in a recent
report that “..an inflammatory process and indirect genotoxic effect by ROS production
seems to be the major mechanism to explain the effects induced by TiO
2
”. However,
SCCS also stated that “a genotoxic effect by direct interaction with DNA cannot be
excluded since TiO
2
was found in the cell nucleus in various in vitro and in vivo studies”
(Scientific Committee on Consumer Safety (SCCS) 2017).
The present working group found that the mechanism of action of the genotoxic and
carcinogenic effects have not been fully clarified (Shi et al. 2013). Secondary
genotoxicity due to particle-induced inflammation is an important and well documented
mechanism of action for the development of lung cancer. However, the available data
did not allow ruling out that TiO
2
NM could also induce cancer through a direct
genotoxic mechanism. Therefore, the present working group considers carcinogenicity as
a non-threshold effect.
31
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
Consequently, the present working group decided to perform the risk assessment based
on both a threshold and a non-threshold mechanism of action.
Cardiovascular effects
NM exposure can lead to cardiovascular effects either: 1. Directly, by translocation of
NMs from the lung to the vascular system. 2. Indirectly, as a consequence of pulmonary
inflammation and acute phase response. 3. Alterations in autonomic nervous system
activity to elicit peripheral effects.
Atherosclerosis is a central cardiovascular effect, which is manifested as increased
plaque deposition or build-up in the arteries. It is initiated by a biological, chemical or
physical insult to the artery walls. Translocated NMs could induce this insult by
interacting directly with the endothelium. This leads to the expression of cell adhesion
molecules (selectins, VCAM-1 and ICAM-1) on the endothelial lining of the arteries,
which facilitates the activation, recruitment and migration of monocytes through the
endothelial monolayer (Hansson and Libby 2006;Cybulsky et al. 2001). Inside the intima
layer, the monocytes differentiate into macrophages and internalize fatty deposits
(mainly oxidized low density lipoprotein), transforming them into foam cells, which is a
major component of the atherosclerotic fatty streaks. The fatty streaks reduce the
elasticity of the artery walls and the foam cells promote a pro-inflammatory environment
by secretion of cytokines and ROS. In addition, foam cells also induce the recruitment of
smooth muscle cells to the intima. Added together, these changes lead to the formation
of plaques on the artery walls. A fibrous cap of collagen and vascular smooth muscle
cells protects the necrotic core and stabilizes the plaque (Libby 2002;Virmani et al. 2005).
Although initially asymptomatic, narrowing of the blood vessels can lead to other
cardiovascular diseases, such as coronary artery disease or stroke. In addition, blood
clots can be formed if the plaque ruptures. These may travel with the bloodstream and
obstruct the blood flow of smaller vessels.
Pulmonary exposure to NMs may also promote accelerated atherosclerosis indirectly
through an induced pulmonary acute phase response. Introduction of NMs to the lung
promotes neutrophil influx and release of pro-inflammatory cytokines, which leads to
increased production of SAA proteins. The SAAs are hydrophobic proteins that upon
secretion in their target organs are able to translocate to the blood. A statistically
significant correlation between Saa3 mRNA levels in the lung and SAA3 protein levels in
the blood have previously been reported (Poulsen et al. 2015a), indicating that SAA3
produced in the target organ translocate to systemic circulation. SAA circulating in the
blood becomes incorporated with HDL, thereby replacing Apolipoprotein A1 (Apo-A1)
as the major HDL-associated protein and forming HDL-SAA. The formation of HDL-
SAA has a double effect on plaque progression: 1.
HDL
is a major component of reverse
cholesterol transport, a multi-stepped process resulting in the movement of cholesterol
through the blood from peripheral tissues (including the artery walls) to the liver. The
formation of SAA-HDL impairs the HDL-mediated reverse cholesterol transport,
resulting in reduced cholesterol transport and an increased systemic total cholesterol
pool (Lindhorst et al. 1997;Steinmetz et al. 1989). 2. SAA and SAA-HDL have been
shown to directly stimulate the transformation of macrophages into foam cells and to
32
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
stimulate uptake of oxidized LDL in the macrophages (Lee et al. 2013). In addition, SAA-
HDL has a lower capacity to promote cellular cholesterol efflux from macrophages than
native HDL (Artl et al. 2000). Pulmonary neutrophil influx has been shown to correlate
with pulmonary Saa3 mRNA levels, SAA3 levels in blood and with deposited surface
area of instilled particles (Saber et al. 2014), which links deposited particle surface area
with biomarkers of risk of developing cardiovascular disease.
In conclusion, the present working group is of the opinion that pulmonary exposure to
particles including TiO
2
NMs can lead to accelerated plaque progression directly,
through translocation, or indirectly, through an induced acute phase response. No single
physicochemical property has been identified as the driver of cardiovascular effects, but
TiO
2
NM surface area a likely important due to the close association with pulmonary
inflammation. As for inflammation, we consider cardiovascular effects as a threshold
effect. This is based on identified dose-response relationships between particle exposure
dose and induced acute phase response (Poulsen et al. 2015a;Saber et al. 2013), and the
close interplay between inflammation, acute phase response and plaque progression.
Dose-response relationships
Inflammation
Strong dose-response relationships have been observed following inhalation (Bermudez
et al. 2004) and intratracheal instillation of TiO
2
NM (Saber et al. 2012a) when dose is
expressed as mass. Inhalation and intratracheal instillation studies have shown that
when rats and mice were exposed TiO
2
, the TiO
2
NM induced a much stronger
pulmonary inflammatory response compared to the same mass of fine TiO
2
particles.
The inflammatory response correlated with the surface area of the deposited particles
irrespectively of size. This dose response relationship has been observed for a number of
low-toxicity, low-solubility particles and it is generally accepted that the inflammatory
response of low toxicity-low solubility particles including TiO
2
is proportional to the
surface area of the deposited particles rather than the mass (reviewed by Oberdörster et
al. (Oberdorster et al. 2005)).
Acute phase response
Strong dose-response relationship has been observed for pulmonary
Saa
mRNA
expression levels in mice intratracheally instilled with TiO
2
NM (Saber et al. 2013).
Saa
mRNA expression levels correlates with neutrophil influx and total deposited surface
area (Saber et al. 2013).
Cancer
As for other low-toxicity low–solubility particles, on a mass basis, the rat tumor response
following pulmonary exposure to ultrafine TiO
2
(Heinrich et al. 1995) is much greater
than for fine TiO
2
(Lee et al. 1985). However, the tumor response in rats exposed to fine
and ultrafine TiO
2
fit on the same dose-response curve when dose is expressed as particle
surface area (NIOSH 2011). This indicates that for the same mass dose of TiO
2
the
tumour response is higher for ultrafine than for fine particles. Based on this, dose-
dependency is assumed for TiO
2
NM-induced lung cancer.
33
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
Particle characteristics
TiO
2
NMs may vary regarding size (and therefore also surface area), crystal form,
coating etc. These are all characteristics that could influence the toxicity. As described
above in the paragraph on dose-response relationship, the surface area of TiO
2
NM is the
best dose predictor for both the inflammatory response and for lung tumors.
TiO
2
exists in different naturally occurring polymorphs including rutile and anatase.
NIOSH concluded that the dose-response relationships for pulmonary inflammation and
lung tumors were not affected by different crystal structures: “The difference in TiO
2
crystal structure in these sub-chronic and chronic studies did not influence the dose-
response relationships for pulmonary inflammation and lung tumors [Bermudez et al.
2002, 2004; Lee et al. 1985; Heinrich et al. 1995]. That is, the particle surface area dose and
response relationships were consistent for the ultrafine (80% anatase, 20% rutile) and fine
(99% rutile) TiO
2
despite the differences in crystal structure.”(NIOSH 2011).
The present working group notes that there is limited available data on the biological
effects of different physico-chemical properties, but the present working group
concludes that the majority of available data support that the surface area (and therefore
also the size) of TiO
2
is a critical driver of particle-induced inflammation and the acute
phase response in the lungs.
34
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
P
REVIOUS RISK ASSESSMENTS OF
T
I
O
2
During the last couple of years, researchers, producers and organizations have proposed
recommended exposure limits (RELs), indicative or derived no-effect-level (INEL/DNEL)
and occupational exposure levels for TiO
2
NM. These have been set based on pulmonary
inflammation or lung cancer. The previous recommendations of exposure limits are
presented below and an overview can be found in table 4.
IARC
In 2006, the International Agency for Research on Cancer (IARC) classified TiO
2
as
possibly carcinogenic to humans (group 2B). This classification was based on sufficient
evidence of carcinogenicity in experimental animals exposed by inhalation and
insufficient evidence in humans. IARC does not differentiate between nano- and fine
particles in their classification (IARC 2010). In Denmark, substances classified as group 1,
2A and 2B by IARC are considered carcinogenic.
ENRHES
One of the first suggestions of a limit value for TiO
2
NM was made within the EU
project ENRHES (Christensen et al. 2011). Due to limited data on TiO
2
NM, the authors
suggest an
indicative
no effect level instead of a
derived
no effect level. The derivation of
an INEL was made under the assumption of a threshold driven mechanism of TiO
2
NM
toxicity: TiO
2
NM induced oxidative stress/inflammation which may result in other
effects such as e.g. cancer.
The INEL 17 µg/m
3
was derived based on the sub-chronic inhalation study of mice, rats
and hamsters by Bermudez et al. (Bermudez et al. 2004). Because rats were the most
sensitive of the tested species, the data from the rats are used for the risk assessment. A
NOAEC (NOAEC
Bermudez
) of 0.5 mg/m
3
was identified for pulmonary influx of
neutrophils immediately after end of exposure in rats exposed 6 hour/day, 5 days/week
for 13 weeks to P25 TiO
2
NM (21nm, 80% anatase/20% rutile).
The calculations of INEL follow the approach given by ECHA (ECHA 2012):
First, the NOAEC
Bermudez
is modified to correct for an 8 hour working day (in Bermudez
et al. (Bermudez et al. 2004) the rats were exposed 6 hour a day) and to correct for a
higher breathing rate in workers (10 m
3
/day) compared to 6.7 m
3
/day at rest:
NOAEC
Corrected
= NOAEC
Bermudez
*6 hour/8 hour * 6.7 m
3
/10 m
3
= 0.25 mg/m
3
Secondly, the corrected NOAEC is adjusted by a number of assessment factors (most of
these are default values suggested by ECHA (ECHA 2012). The following assessment
factors are used. To adjust for interspecies extrapolation, an assessment factor of 1.5 was
used (default factor is 2.5) because the observed toxic effects do not involve metabolism
and therefore there is no need for allometric scaling):
35
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
Interspecies extrapolation (default factor is 2.5):
Intraspecies interpolation (default factor for workers):
Extrapolation from sub-chronic to chronic (default factor):
The overall assessment factor,
AF
Total
= 1.5 * 5 * 2 = 15
This results in an INEL for chronic inhalation for pulmonary inflammation of:
INEL = NOAEC
Corrected
/AF
Total
= 0.25 mg/m
3
/ 15 = 0.017 mg/m
3
= 17 µg/m
3
1.5
5
2
NEDO
The NEDO project also used the sub-chronic inhalation study by Bermudez et al.
(Bermudez et al. 2004) as basis for calculation (Nakanishi and Gamo 2011). However, in
contrast to Christensen et al. (Christensen et al. 2011), Nakanishi and Gamo chose to use
a NOAEC (NOAEC
Bermudez
) of 2.0 mg/m
3
for pulmonary influx of neutrophils in rats.
From 4 weeks after end of exposure and the following time points the NOAEC is 2.0
mg/m
3
, while the NOAEC used by Christensen et al (Christensen et al. 2011) is the
NOAEC immediately after end of exposure (please see table 2 for details, paragraph on
subacute studies).
The corrected NOAEC for human exposure is calculated to be 1.8 mg/m
3
.
The assessment factor is: AF=3
This results in the following recommendation by Nakanishi and Gamo, 2011:
OEL = NOAEC
corrected
/AF= 1.8 mg/m
3
/3= 0.61 mg/m
3
NIOSH
NIOSH suggested the following recommended airborne exposure limits (as time-
weighted average (TWA) concentrations for up to 10 hr/day during a 40-hour week): 0.3
mg/m
3
for ultrafine (including engineered nanoscale) TiO
2
and 2.4 mg/m
3
for fine TiO
2
.
“These recommendations represent levels that over a working lifetime are estimated to
reduce risks of lung cancer to below 1 in 1,000. The recommendations are based on using
chronic inhalation studies in rats to predict lung tumor risks in humans.”(NIOSH 2011).
NIOSH also derived exposure concentrations that are designed to prevent pulmonary
inflammation. These are 0.004 mg/m
3
for ultrafine TiO
2
and 0.04 mg/m
3
for fine TiO
2
.
These were derived based on a benchmark dose analysis for pulmonary inflammation in
rats followed by an extrapolation of the rat benchmark doses to humans. The starting
points for the calculations were 0.9 mg/m
3
and 0.11 mg/m
3
for the fine and ultrafine TiO
2
,
respectively. Compared to the RELs accepting a risk of cancer below 1 out of 1,000 there
would be a zero excess risk of cancer development due to secondary toxicity at exposure
limits preventing pulmonary inflammation.
NIOSH showed that total deposited particle surface area of TiO
2
particles of different
sizes (fine and ultrafine) and different crystal structure (80% anatase/20% rutile and 99%
36
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
rutile) can explain the observed variation in TiO
2
particle-induced pulmonary
inflammation and lung cancer in rat inhalation studies: “…when rats were exposed to
TiO
2
in sub-chronic inhalation studies, no difference in pulmonary inflammation
response to fine and ultrafine TiO
2
particles of different crystal structure (i.e., 99% rutile
vs. 80% anatase/20% rutile) was observed once dose was adjusted for particle surface
area [Bermudez et al. 2002, 2004]; nor was there a difference in the lung tumor response
in the chronic inhalation studies in rats at a given surface area dose of these fine and
ultrafine particles (i.e., 99% rutile vs. 80% anatase/20% rutile) [Lee et al. 1985; Heinrich et
al. 1995]. Therefore, NIOSH concludes that the scientific evidence supports surface area
as the critical metric for occupational inhalation exposure to TiO
2
.”(NIOSH 2011).
Scaffold project
Recently, a recommendation of a limit value for TiO
2
NM was made within the frames of
the EU project Scaffold and was published by Stockmann-Juvala et al.(Stockmann-Juvela
et al. 2014).The Scaffold project identified pulmonary inflammation as the critical health
effect for TiO
2
. Similar to Christensen et al. (Christensen et al. 2011) and the NEDO
project (Nakanishi and Gamo 2011), the project uses the sub-chronic inhalation study by
Bermudez et al. (Bermudez et al. 2004) as basis for the calculation. Similar to Christensen
et al. 2011, Stockmann-Juvala et al., 2014 uses a NOAEC (NOAEC
Bermudez
) of 0.5 mg/m
3
for
pulmonary influx of neutrophils in rats immediately after end of exposure is chosen as
starting point for the calculations.
First, the NOAEC
Bermudez
is modified to correct for an 8 hour working day (in Bermudez
et al. (Bermudez et al. 2004) the rats were exposed 6 hour a day) and to correct for a
higher breathing rate in workers (10 m
3
/day) compared to 6.7 m
3
/day at rest:
NOAEC
Corrected
= NOAEC
Bermudez
*6 hour/8 hour * 6.7 m
3
/10 m
3
= 0.25 mg/m
3
Secondly, the corrected NOAEC is adjusted by an assessment factor to take differences
between sensitivity between individuals into account:
AF
= 2.5
This results in an OEL for nanoTiO
2
:
OEL = NOAEC
Corrected
/AF = 0.25 mg/m
3
/ 2.5 = 0.1 mg/m
3
= 100 µg/m
3
No data were identified for the determination of an OEL for dermal exposure.
ECHA’s Committee for Risk Assessment (RAC)
ECHA’s Committee for Risk Assessment (RAC) has concluded that the available
scientific evidence meets the criteria in the CLP Regulation to classify TiO
2
as a substance
suspected of causing cancer through the inhalation route (RAC, 2017).
37
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
Summary of the evaluations
As shown in table 4, three of the recommendations use the same approach on the results
from the same sub-chronic rat inhalation study (Bermudez et al. 2004). However, due to
different choice of starting point and/or different assessment factors the derived
recommendations are in the range from 17 µg/m
3
– 610 µg/m
3
(Christensen et al.
2011;Stockmann-Juvela et al. 2014;Nakanishi and Gamo 2011). Based on a benchmark
dose approach NIOSH suggested the following recommended airborne exposure limits
(as time-weighted average (TWA) concentrations for up to 10 hr/day during a 40-hour
week): 0.3 mg/m
3
for ultrafine (including engineered nanoscale) TiO
2
and 2.4 mg/m
3
for
fine TiO
2
. “These recommendations represent levels that over a working lifetime are
estimated to reduce risks of lung cancer to below 1 in 1,000. The recommendations are
based on using chronic inhalation studies in rats to predict lung tumor risks in
humans.”(NIOSH 2011)
38
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
2130137_0041.png
Table 4. Overview of suggested OELs for TiO
2
NM by different organizations/researchers
Methodology for OEL development and reference/project
ENRHES
NEDO
NIOSH
Scaffold
(Christensen et
(NIOSH 2011)
(Stockmann-Juvela
(
Nakanishi
and
al. 2011)
et al. 2014)
Gamo 2011)
Critical effect
Pulmonary
Pulmonary
Lung cancer
a
Pulmonary inflammation
a
Pulmonary
inflammation
inflammation
inflammation
Key study
(Bermudez et al.
(Bermudez et al.
(Lee et al. 1985;Muhle et
(Bermudez et al.
(Bermudez et al.
2004)
2004)
al. 1991;Heinrich et al.
2002;Bermudez et al.
2004)
1995)
2004;Cullen et al.
2002;Tran et al. 1999)
Risk determinant
NOAEC
NOAEC
BMDL
BMD
NOAEC
associated with 1/1000
Particle surface area per
excess risk of cancer
gram of lung tissue
associated with 4%
inflammatory response of
neutrophils
Risk level in
0.5 mg/m
3
2 mg/m
3
0.5 mg/m
3
rodents
Corrected starting
0.25 mg/m
3a
0.29 mg/m
3
0.11 mg/m
3
0.25 mg/m
3
point
Uncertainty factors
Interspecies
Intraspecies
Sub-chronic
to chronic
Overall uncertainty
factor
Suggested OEL
a
1.5
5
2
15
0.017 mg/m
3
(17 µg/m
3
)
3
1
1
3
0.61 mg/m
3
(610 µg/m
3
)
0.3 mg/m
3
(300 µg/m
3
)
2.5
10
-
25
0.004 mg/m
3
(4 µg/m
3
)
1
2.5
1
2.5
0.1 mg/m
3
(100 µg/m
3
)
NIOSH calculated exposure limits based on both pulmonary inflammation and lung cancer. However, NIOSH’s final recommendation is based
on lung cancer rather than pulmonary inflammations. For transparency, both results are shown in the table.
39
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
S
CIENTIFIC BASIS FOR SETTING AN OCCUPATIONAL
EXPOSURE LIMIT
Different methods exist for calculating OELs. The choice of method depends on the
mode of action of the substance, and can fundamentally be split up in two approaches:
Threshold effects or non-threshold effects. The threshold effect approach relies on the
assumption that the organism can withstand a certain dose before adverse effects occur,
whereas for non-threshold effects it is assumed that any exposure to the substance can
result in adverse effects. In this report, we will calculate proposed OELs based both on
threshold effects and non-threshold effects.
Endpoint: Inflammation
Pulmonary inflammation is a defense mechanism when particles or other types of
foreign material enter the lungs. Particle-induced pulmonary inflammation is considered
to be one of the key steps leading to lung cancer by a secondary genotoxic mechanism
(NIOSH 2011). Furthermore there is a close interplay between inflammation, the acute
phase response and cardiovascular plaque progression (Poulsen et al. 2015a;Saber et al.
2013). Thus, the derivation of a DNEL based on inflammation has been made under the
assumption of a threshold-driven mechanism of TiO
2
NM toxicity: TiO
2
NM induced
oxidative stress/inflammation which may result in other effects such as e.g. cancer and
cardiovascular disease.
Our approach for an OEL for TiO
2
NM follows the traditional approach for setting
health-based OELs: 1) identification of critical effect, 2) identification of the NOAEC, 3)
calculation of OEL using assessment factors adjusting for inter and intra species
differences).
In the current report we use the DNEL as recommended by ECHA as the OEL for
toxicological effects having thresholds (ECHA 2012).
The DNEL of 10 µg/m
3
is derived based on the sub-chronic inhalation study of mice, rats
and hamsters by Bermudez et al. (Bermudez et al. 2004). Rats were the most sensitive of
the tested species, and the data from the rats are used for the DNEL derivation. A
NOAEC (NOAEC
Bermudez
) of 0.5 mg/m
3
was identified for pulmonary influx of
neutrophils immediately after end of exposure in rats exposed 6 hour/day, 5 days/week
for 13 weeks to P25 TiO
2
NM (21nm, 80% anatase/20% rutile). Histopathological changes
in the lungs were dose and time dependent.
The study by Bermudez et al (Bermudez et al. 2004) is the only sub-chronic dose-
response inhalation study with TiO
2
NM identified. In addition to this study we have
identified a range of inhalation studies of shorter exposure time using TiO
2
particles
(Ma-Hock et al. 2009;Noel et al. 2012;Rossi et al. 2010a;Rossi et al. 2010b;Baisch et al.
2014;Lindberg et al. 2012). Overall they support that a NOAEC level is in the range of
0.5-2 mg/m
3
.
The calculations of the DNEL follow the approach as set out in the REACH guidance
(ECHA 2012):
40
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
First, the NOAEC
Bermudez
is modified to correct for an 8 hour working day (in Bermudez
et al. (Bermudez et al. 2004)) the rats were exposed 6 hour a day) and to correct for a
higher breathing rate in workers at light work (10 m
3
/day) compared to 6.7 m
3
/day at
rest:
NOAEC
Corrected
= NOAEC
Bermudez
*6 hour/8 hour * 6.7 m
3
/10 m
3
= 0.25 mg/m
3
Secondly, the corrected NOAEC is adjusted by a number of assessment factors (most of
these are default values suggested by ECHA.
Inflammation is considered an acute response. Due to the accumulation of particles over
time, we have chosen to use the default assessment factor 2 to extrapolate from sub-
chronic to chronic exposure. The following default assessment factors are used:
Interspecies extrapolation:
Intraspecies interpolation (default factor for workers):
Extrapolation from sub-chronic to chronic:
The overall assessment factor (AF
Total
),
AF
Total
= 2.5 * 5 * 2 = 25
This results in a DNEL for chronic inhalation for pulmonary inflammation of:
DNEL = NOAEC
Corrected
/AF
Total
= 0.25 mg/m
3
/ 25 = 0.01 mg/m
3
= 10 µg/m
3
2.5
5
2
Endpoint: Cancer
The present working group has chosen not to use the epidemiological study by Ellis et
al. (Ellis et al. 2013) as basis for an OEL suggestion for several reasons, including the lack
of information on particle size, lack of dose-response relationship between lung cancer
incidence and cumulative TiO
2
dose, lack of description of the reference groups
including information on exposure, lack of information about the lung cancer incidence
in the reference groups.
Instead, the derivation of an OEL based on cancer has been made under the assumption
of a non-threshold driven mechanism of TiO
2
NM toxicity.
The OEL is derived based on the chronic inhalation study of mice and rats by Heinrich et
al. (Heinrich et al. 1995). Lung tumor rate in mice exposed to TiO
2
was not statistically
different from the lung tumor rate in mice exposed to filtered air. Therefore, as the most
sensitive of the tested species, data from the rats are used for the risk assessment.
The lowest effect level for lung cancer was observed in rats, where increased lung cancer
incidence was found at 10 mg/m
3
. Lung cancer incidence in TiO
2
exposed rats was 32%
(32/100), while the cancer incidence in control rats was 0.5% (1/217).
41
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
2130137_0044.png
Increased lung cancer incidence was observed in rats at 10 mg/m
3
. Both malignant and
non-malignant tumors were included in accordance with the REACH guideline stating
that: “malignant tumours as well as benign tumours that are suspected of possibly
progressing to malignant tumours are taken into account in obtaining the dose-
descriptor values” (ECHA, 2012).
Table 5. Cancer incidence and TiO
2
lung burden at different TiO
2
mass air concentrations in a
chronic inhalation study (Heinrich et al. 1995)
.
Total cancer
incidence
TiO
2
lung
burden
(mg/lung)
0 mg/m
3
1/217
10 mg/m
3
32/100
39
Observed excess cancer incidence at 10 mg/m
3
:
(32/100- 1/217)/(1-1/217)= 0.32 =32 %
Method I
The present working group has chosen to use the approach used by Kasai et al (Kasai et
al. 2016) and Erdely et al (Erdely et al. 2013), who use the measured lung burden in rats
exposed by inhalation and the alveolar surface area of rats and humans to estimate the
human equivalent lung burden:
At 10 mg/m
3
, the amount of pulmonary deposited TiO
2
after 2 years of inhalation was
determined to be 39 mg/rat lung (Heinrich et al. 1995).
Human lung burden equals:
Rat lung burden (39 mg) × Human alveolar surface area (102 m
2
) / rat alveolar surface
area (0.4 m
2
) = 9945 mg per human lung.
We assume using standard values that human ventilation is 20 L/min during light work
(1.2 m
3
/h), work related exposure for 8 h per day, 5 days per week, 45 working weeks per
year, over a working lifetime of 45 years. The deposition rate was not reported to in the
Heinrich study. For the calculation, we have used a deposition of 8.6% based on an
inhalation study by (Hougaard et al. 2010). In that study, mice were exposed by
inhalation 1h/day for 11 days to 42 mg/m
3
aerosolized powder of rutile TiO
2
with an
average crystallite size of 21 nm. The pulmonary deposition fraction was estimated to be
8.6% based on the observed particle size distribution in the aerosol.
A lung burden of 9945 mg in humans would require that workers are exposed:
Air concentration = 9945 mg/(8h/day x 5 day/week x 45 weeks/year x 45 years x 1.2 m
3
/h
x 0.086) = 1.2 mg/m
3
.
42
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
2130137_0045.png
Thus, at an air concentration of 1.2 mg/m
3
during a 45 year work life, an excess lung
cancer incidence of 32% is expected.
Assuming a linear dose-response relationship, then 1% excess lung cancer is expected at
(1.2 mg/m
3
)/32 = 0.04 mg/m
3
(40 µg/m
3
).
The TiO
2
NM air concentrations resulting in different excess lung cancer incidences are
given in the table below.
Table 6. Calculated excess lung cancer incidences at different TiO
2
NM mass concentrations
based on method I.
Excess lung cancer
incidence
1:1 000
1: 10 000
1: 100 000
TiO
2
NM Air
concentration (µg/m
3
)
4
0.4
0.04
Method II
ECHA (ECHA 2012; SCHER/SCCP/SCENIHR 2009), calculated based on the two year
TiO
2
NM inhalation study in rats by (Heinrich et al. 1995) (Table 5):
Excess cancer risk:
Observed excess cancer incidence at 10 mg/m
3
:
(32/100- 1/217)/(1-1/217)= 0.32 =32 %
Correction of dose metric for humans during occupational exposure (8h/d):
10 mg/m
3
x (18 h/day)/(8 h/day) x (6.7 m
2
/10 m
2
) = 15 mg/m
3
Calculation of unit risk for cancer:
Risk level = exposure level x unit risk
0.32 = 15 000 µg/m
3
x unit risk
Unit risk = 2.1 x 10
-5
per µg/m
3
At a dose of 1 µg/m
3
, 2.1 x 10
-5
excess cancers are expected.
Calculation of dose levels corresponding to risk level of 10
-5
(and other risk levels)
10
-5
risk level = exposure level x unit risk (2.1 x 10
-5
per µg/m
3
)
Exposure level (10
-5
) = 0.47 µg/m
3
Thus, at 0.47 µg/m
3
, 1:100 000 excess lung cancer cases can be expected.
43
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
2130137_0046.png
Table 7. Calculated excess lung cancer incidence at different TiO
2
NM mass concentrations
based on method II.
Excess lung cancer
incidence
1:1,000
1: 10,000
1: 100,000
TiO
2
NM Air
concentration (µg/m
3
)
47
4.7
0.47
44
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
C
ONCLUSION
The present working group evaluated the relevant literature on TiO
2
NM from both
epidemiological and animal inhalation studies. None of the identified epidemiological
studies provided information on the particle size range of the TiO
2
, thus making it
impossible to determine whether the exposures included TiO
2
NM. Therefore it was
decided to base the suggested health-based OEL on data from experimental animal
studies.
Pulmonary inflammation and carcinogenicity was observed in sub-chronic and chronic
inhalation studies in rats. The present working group regards inflammation and
carcinogenicity as the critical adverse effects and the subsequent risk assessments are
conducted based on studies reporting these effects. TiO
2
NM induced cardiovascular
effects were also identified in animal studies. But as none of these studies were sub-
chronic or chronic inhalation studies, they were not suitable for risk assessment.
However, the present working group regards the acute phase response as a biomarker of
cardiovascular effects. Due to the close association between pulmonary inflammation
and the acute phase response, the present working group regards inflammation as a
proxy for cardiovascular effects.
The present working group found strong dose response relationships for neutrophil
influx as a marker of pulmonary inflammation (Bermudez et al. 2004). Neutrophil influx
was predicted by deposited surface area. The working group considers inflammation as
a threshold effect.
The present working group found that the mechanism of action of the carcinogenic effect
has not been fully clarified. Secondary genotoxicity due to particle-induced
inflammation is an important and well documented mechanism of action for the
development of lung cancer. However, the available data did not allow ruling out that
TiO
2
NM could also induce cancer through a direct genotoxic mechanism. Therefore, the
present working group considers carcinogenicity as a non-threshold effect.
Consequently, the present working group decided to perform the risk assessment based
on both a threshold and a non-threshold mechanism of action.
The working group considered that data from two rat inhalation studies were the best
basis for risk assessment. The following studies were selected to be used for calculation
of DNEL and excess cancer risk, respectively: A 13 week sub-chronic inhalation study in
rats (0, 0.5, 2.0 and 10 mg/m
3
) (Bermudez et al. 2004) and a 2 year chronic cancer
inhalation study in rats (0 and 10 mg/m
3
) (Heinrich et al. 1995). Table 8 shows a DNEL
for pulmonary inflammation derived based on the sub-chronic inhalation study of rats as
the most sensitive of three tested species, and exess lung cancer risk at 1 in 1 000, 1 in 10
000 and 1 in 100 000 derived using two different approaches.
45
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
2130137_0048.png
Table 8. Overview of DNEL based on a threshold based mechanism of action and exposure
levels resulting in extra cancer risk levels at 1:1000, 1:10 000 and 1: 100 000 based on a non-
threshold based mechanism of action using two different approaches.
Threshold
based
Non-threshold
based
DNEL
Extra cancer
risk
1:1000
1:10 000
1:100 000
Suggestion of an OEL for TiO
2
NM
Inflammation
Lung Cancer
Lung cancer
(method I)
(method II)
10 µg/m
3
4 µg/m
3
0.4 µg/m
3
0.04 µg/m
3
47
4.7
0.47 µg/m
3
Both of studies used for the risk assessment used P25 TiO
2
NM (15-40 nm diameter, 80%
anatase/20% rutile). TiO
2
NMs differ regarding size and surface area but also coating,
shape, crystal structure etc. The present working group notes that there is limited
available data on the biological effects of different physico-chemical properties, but the
present working group concludes that the majority of available data support that the
surface area (and therefore the size) of TiO
2
is a critical driver of particle-induced
inflammation and the acute phase response in the lungs.
The present working group also notes that NIOSH showed that particle surface area of
TiO
2
particles of different sizes (fine and ultrafine) and different crystal structure (80%
anatase/20% rutile and 99% rutile) can explain the observed variation in TiO
2
particle-
induced pulmonary inflammation and lung cancer in rat inhalation studies. This stresses
the importance of the surface area as a predictor for the inflammatory and carcinogenic
response.
The present working group regards cancer as the most critical effect.
The DNEL approach relies heavily on the assumption of a threshold effect on
inflammation and carcinogenicity. The present working group is of the opinion that
there is still uncertainly whether this is the case for TiO
2
NM
–induced
carcinogenicity.
Two different approaches were used for calculating excess lung cancer risk based on the
same chronic inhalation study. In the first approach, lung burden in rats after two years
of exposure was used to estimate the exposure levels for occupational exposure. In the
second approach, air concentrations were used directly. Independently of the applied
method for risk assessment, the resulting exposure levels were all very low. These levels
are all more than 100-fold lower than the current Danish OEL for titanium of 6 mg/m
3
(measured as Ti, corresponding to 10 mg/m
3
for TiO
2
).
The present working group recommends the approach using the excess lung cancer risk
estimates based on lung burden, since this approach takes the actual retained pulmonary
dose into account. Thus, the expected excess lung cancer risk based on lung burden
approach is 1:1 000 at 4 µg/m
3
, 1:10 000 at 0.4 µg/m
3
and 1:100 000 at 0.04 µg/m
3
TiO
2
NM.
46
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
R
EFERENCES
American Cancer Society. Lifetime risk of developing or dying from cancer. American
Cancer Society, 2018.
https://www.cancer.org/cancer/cancer-basics/lifetime-probability-of-developing-or-
dying-from-cancer.html
Artl A, Marsche G, Lestavel S, Sattler W, Malle E. Role of serum amyloid A during
metabolism of acute-phase HDL by macrophages. Arterioscler Thromb Vasc Biol
2000;20:763-772.
Baisch BL, Corson NM, Wade-Mercer P, Gelein R, Kennell AJ, Oberdörster G, Elder A.
Equivalent titanium dioxide nanoparticle deposition by intratracheal instillation and
whole body inhalation: The effect of dose rate on acute respiratory tract inflammation.
Part Fibre Toxicol 2014;11:5.
Bermudez E, Mangum JB, Asgharian B, Wong BA, Reverdy EE, Janszen DB, Hext PM,
Warheit DB, Everitt JI. Long-term pulmonary responses of three laboratory rodent
species to subchronic inhalation of pigmentary titanium dioxide particles. Toxicol Sci
2002;70:86-97.
Bermudez E, Mangum JB, Wong BA, Asgharian B, Hext PM, Warheit DB, Everitt JI.
Pulmonary responses of mice, rats, and hamsters to subchronic inhalation of ultrafine
titanium dioxide particles. Toxicol Sci 2004;77:347-357.
Boffetta P, Gaborieau V, Nadon L, Parent ME, Weiderpass E, Siemiatycki J. Exposure to
titanium dioxide and risk of lung cancer in a population-based study from Montreal.
Scand J Work Environ Health 2001;27:227-232.
Boffetta P, Soutar A, Cherrie JW, Granath F, Andersen A, Anttila A, Blettner M,
Gaborieau V, Klug SJ, Langard S, Luce D, Merletti F, Miller B, Mirabelli D, Pukkala E,
Adami HO, Weiderpass E. Mortality among workers employed in the titanium dioxide
production industry in Europe. Cancer Causes Control 2004;15:697-706.
Boisen AM, Shipley T, Jackson P, Hougaard KS, Wallin H, Yauk CL, Vogel U.
NanoTIO(2) (UV-Titan) does not induce ESTR mutations in the germline of prenatally
exposed female mice. Part Fibre Toxicol 2012;9:19.
Bourdon JA, Saber AT, Jacobsen NR, Jensen KA, Madsen AM, Lamson JS, Wallin H,
Møller P, Loft S, Yauk CL, Vogel UB. Carbon black nanoparticle instillation induces
sustained inflammation and genotoxicity in mouse lung and liver. Part Fibre Toxicol
2012;9:5.
Cancer Risks UK. Worldwide cancer mortality statistics. Cancer Risks UK. 2018.
https://www.cancerresearchuk.org/health-professional/cancer-statistics/worldwide-
cancer/mortality
Chen JL, Fayerweather WE. Epidemiologic study of workers exposed to titanium-
dioxide. J Occup Environ Med 1988;30:937-942.
47
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
Chen T, Hu J, Chen C, Pu J, Cui X, Jia G. Cardiovascular effects of pulmonary exposure
to titanium dioxide nanoparticles in ApoE knockout mice. J Nanosci Nanotechnol
2013;13:3214-3222.
Christensen FM, Johnston HJ, Stone V, Aitken RJ, Hankin S, Peters S, Aschberger K.
Nano-TiO
2
--feasibility and challenges for human health risk assessment based on open
literature. Nanotoxicology 2011; 5:110-124.
Cullen RT, Jones AD, Miller BG, Tran CL, Davis JMG, Donaldson K, Wilson M, Stone V,
Morgan A. Toxicity of volcanic ash from Montserrat. Research Report TM/02/01.
Edingburgh: Institute of Occupational Medicine, 2002.
Cybulsky MI, Iiyama K, Li H, Zhu S, Chen M, Iiyama M, Davis V, Gutierrez-Ramos JC,
Connelly PW, Milstone DS. A major role for VCAM-1, but not ICAM-1, in early
atherosclerosis. J Clin Invest 2001;107:1255-1262.
Debia M, Bakhiyi B, Ostiguy C, Verbeek JH, Brouwer DH, Murashov V. A systematic
review of reported exposure to engineered nanomaterials. Ann Occup Hyg 2016;60:916-
935.
ECHA. Guidance on information requirements and chemical safety assessment.
Appendix R8-15 Recommendations for nanomaterials applicable to Chapter R.8
Characterisation of dose [concentration] - response for human health. ECHA-12-G-09-
EN. European Chemicals Agency, 2012.
ECHA. Guidance on information requirements and chemical safety assessment Chapter
R.8: Characterisation of dose [concentration]-response for human health. ECHA-2010-G-
19-EN. European Chemicals Agency, 2012.
Ellis ED, Watkins J, Tankersley W, Phillips J, Girardi D. Mortality among titanium
dioxide workers at three DuPont plants. J Occup Environ Med 2010;52:303-309.
Ellis ED, Watkins JP, Tankersley WG, Phillips JA, Girardi DJ. Occupational exposure and
mortality among workers at three titanium dioxide plants. Am J Ind Med 2013;56:282-
291.
Engler-Chiurazzi EB, Stapleton PA, Stalnaker JJ, Ren X, Hu H, Nurkiewicz TR, McBride
CR, Yi J, Engels K, Simpkins JW. Impacts of prenatal nanomaterial exposure on male
adult Sprague-Dawley rat behavior and cognition. J Toxicol Environ Health A
2016;79:447-452.
Erdely A, Dahm M, Chen BT, Zeidler-Erdely PC, Fernback JE, Birch ME, Evans DE,
Kashon ML, Deddens JA, Hulderman T, Bilgesu SA, Battelli L, Schwegler-Berry D,
Leonard HD, McKinney W, Frazer DG, Antonini JM, Porter DW, Castranova V,
Schubauer-Berigan MK. Carbon nanotube dosimetry: from workplace exposure
assessment to inhalation toxicology. Part Fibre Toxicol 2013;10:53.
48
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
European Commission. Definition of a nanomaterial. Last update: 22-2-2017. European
Commission, 2018.
http://ec.europa.eu/environment/chemicals/nanotech/faq/definition_en.htm
Ferin J, Oberdörster G, Penney DP. Pulmonary Retention of Ultrafine and Fine Particles
in Rats. Am J Respir Cell Mol Biol 1992;6:535-542.
Fryzek JP, Chadda B, Marano D, White K, Schweitzer S, McLaughlin JK, Blot WJ. A
cohort mortality study among titanium dioxide manufacturing workers in the United
States. J Occup Environ Med 2003;45:400-409.
Gabay C, Kushner I. Acute-phase proteins and other systemic responses to
inflammation. N Engl J Med 1999;340:448-454.
Geiser M, Casaulta M, Kupferschmid B, Schulz H, Semmler-Behnke M, Kreyling W. The
role of macrophages in the clearance of inhaled ultrafine titanium dioxide particles.
Am J Respir Cell Mol Biol 2008;38:371-376.
Geiser M, Kreyling WG. Deposition and biokinetics of inhaled nanoparticles.
Part Fibre Toxicol 2010;7:2.
Haberl N, Hirn S, Holzer M, Zuchtriegel G, Rehberg M, Krombach F. Effects of acute
systemic administration of TiO2, ZnO, SiO2, and Ag nanoparticles on hemodynamics,
hemostasis and leukocyte recruitment. Nanotoxicology 2015;9:963-971.
Halappanavar S, Jackson P, Williams A, Jensen KA, Hougaard KS, Vogel U, Yauk CL,
Wallin H. Pulmonary response to surface-coated nanotitanium dioxide particles includes
induction of acute phase response genes, inflammatory cascades, and changes in
microRNAs: A toxicogenomic study. Environ Mol Mutagen 2011;52:425-439.
Hansson GK, Libby P. The immune response in atherosclerosis: A double-edged sword.
Nat Rev Immunol 2006;6:508-519.
Hathaway QA, Nichols CE, Shepherd DL, Stapleton PA, McLaughlin SL, Stricker JC,
Rellick SL, Pinti MV, Abukabda AB, McBride CR, Yi J, Stine SM, Nurkiewicz TR,
Hollander JM. Maternal-engineered nanomaterial exposure disrupts progeny cardiac
function and bioenergetics. Am J Physiol Heart Circ Physiol 2017;312:H446-H458.
Heinrich U, Fuhst R, Rittinghausen S, Creutzenberg O, Bellmann B, Koch W, Levsen K.
Chronic inhalation exposure of wistar rats and two different strains of mice to diesel
engine exhaust, carbon black, and titanium dioxide. Inhal Toxicol 1995;7:533-556.
Hougaard KS, Jackson P, Jensen KA, Sloth JJ, Löschner K, Larsen EH, Birkedal RK,
Vibenholt A, Boisen AM, Wallin H, Vogel U. Effects of prenatal exposure to surface-
coated nanosized titanium dioxide (UV-Titan). A study in mice. Part Fibre Toxicol
2010;7:16.
49
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
Husain M, Saber AT, Guo C, Jacobsen NR, Jensen KA, Yauk CL, Williams A, Vogel U,
Wallin H, Halappanavar S. Pulmonary instillation of low doses of titanium dioxide
nanoparticles in mice leads to particle retention and gene expression changes in the
absence of inflammation. Toxicol Appl Pharmacol 2013;269:250-262.
Husain M, Wu D, Saber AT, Decan N, Jacobsen NR, Williams A, Yauk CL, Wallin H,
Vogel U, Halappanavar S. Intratracheally instilled titanium dioxide nanoparticles
translocate to heart and liver and activate complement cascade in the heart of C57BL/6
mice. Nanotoxicology 2015;9:1013-1022.
IARC. Carbon Black, Titanium Dioxide, and Talc. IARC Working Group on the
Evaluation of Carcinogenic Risks to Humans. IARC Monographs on the Evaluation of
Carcinogenic Risks to Humans, 93. Lyon: WHO, The International Agency for Research
on Cancer, 2010.
Jackson P, Halappanavar S, Hougaard KS, Williams A, Madsen AM, Lamson JS,
Andersen O, Yauk C, Wallin H, Vogel U. Maternal inhalation of surface-coated
nanosized titanium dioxide (UV-Titan) in C57BL/6 mice: Effects in prenatally exposed
offspring on hepatic DNA damage and gene expression. Nanotoxicology 2013;7:85-96.
Johnson BD, Kip KE, Marroquin OC, Ridker PM, Kelsey SF, Shaw LJ, Pepine CJ, Sharaf
B, Bairey Merz CN, Sopko G, Olson MB, Reis SE. Serum amyloid A as a predictor of
coronary artery disease and cardiovascular outcome in women: The National Heart,
Lung, and Blood Institute-Sponsored Women's Ischemia Syndrome Evaluation (WISE).
Circulation 2004;109:726-732.
Kasai T, Umeda Y, Ohnishi M, Mine T, Kondo H, Takeuchi T, Matsumoto M, Fukushima
S. Lung carcinogenicity of inhaled multiwalled carbon nanotube in rats.
Part Fibre Toxicol 2016;13:53.
Kinaret P, Ilves M, Fortino V, Rydman E, Karisola P, Lahde A, Koivisto J, Jokiniemi J,
Wolff H, Savolainen K, Greco D, Alenius H. Inhalation and oropharyngeal aspiration
exposure to rod-like carbon nanotubes induce similar airway inflammation and
biological responses in mouse lungs. ACS Nano 2017;11:291-303.
Koivisto AJ, Lyyranen J, Auvinen A, Vanhala E, Hameri K, Tuomi T, Jokiniemi J.
Industrial worker exposure to airborne particles during the packing of pigment and
nanoscale titanium dioxide. Inhal Toxicol 2012;24:839-849.
Kreyling WG, Holzwarth U, Haberl N, Kozempel J, Hirn S, Wenk A, Schleh C, Schaffler
M, Lipka J, Semmler-Behnke M, Gibson N. 2017a. Quantitative biokinetics of titanium
dioxide nanoparticles after intravenous injection in rats: Part 1. Nanotoxicology
2017a;11:434-442.
Kreyling WG, Holzwarth U, Haberl N, Kozempel J, Wenk A, Hirn S, Schleh C, Schaffler
M, Lipka J, Semmler-Behnke M, Gibson N. Quantitative biokinetics of titanium dioxide
nanoparticles after intratracheal instillation in rats: Part 3. Nanotoxicology 2017b;11:454-
464.
50
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
Kreyling WG, Holzwarth U, Schleh C, Kozempel J, Wenk A, Haberl N, Hirn S, Schaffler
M, Lipka J, Semmler-Behnke M, Gibson N. Quantitative biokinetics of titanium dioxide
nanoparticles after oral application in rats: Part 2. Nanotoxicology 2017c;11:443-453.
Kwon S, Yang YS, Yang HS, Lee J, Kang MS, Lee BS, Lee K, Song CW. Nasal and
pulmonary toxicity of titanium dioxide nanoparticles in rats. Toxicol Res 2012;28:217-224.
Kyjovska ZO, Boisen AM, Jackson P, Wallin H, Vogel U, Hougaard KS. Daily sperm
production: Application in studies of prenatal exposure to nanoparticles in mice.
Reprod Toxicol 2013;36:88-97.
Lee HY, Kim SD, Baek SH, Choi JH, Cho KH, Zabel BA, Bae YS. Serum amyloid A
stimulates macrophage foam cell formation via lectin-like oxidized low-density
lipoprotein receptor 1 upregulation. Biochem Biophys Res Commun 2013;433:18-23.
Lee KP, Trochimowicz HJ, Reinhardt CF. Pulmonary response of rats exposed to
titanium dioxide (TiO2) by inhalation for two years. Toxicol Appl Pharmacol
1985;79:179-192.
Libby P. Inflammation in atherosclerosis. Nature 2002;420:868-874.
Lindberg HK, Falck GC, Catalán J, Koivisto AJ, Suhonen S, Järventaus H, Rossi EM,
Nykäsenoja H, Peltonen Y, Moreno C, Alenius H, Tuomi T, Savolainen KM, Norppa H.
Genotoxicity of inhaled nanosized TiO(2) in mice. Mutat Res 2012;745:58-64.
Lindhorst E, Young D, Bagshaw W, Hyland M, Kisilevsky R. Acute inflammation, acute
phase serum amyloid A and cholesterol metabolism in the mouse. Biochim Biophys Acta
1997;1339:143-154.
Lowe GD. The relationship between infection, inflammation, and cardiovascular disease:
an overview. Ann Periodontol 2001;6:1-8.
Ma-Hock L, Burkhardt S, Strauss V, Gamer AO, Wiench K, van Ravenzwaay B,
Landsiedel R. Development of a short-term inhalation test in the rat using nano-titanium
dioxide as a model substance. Inhal Toxicol 2009;21:102-118.
Mezaki T, Matsubara T, Hori T, Higuchi K, Nakamura A, Nakagawa I, Imai S, Ozaki K,
Tsuchida K, Nasuno A, Tanaka T, Kubota K, Nakano M, Miida T, Aizawa Y. Plasma
levels of soluble thrombomodulin, C-reactive protein, and serum amyloid A protein in
the atherosclerotic coronary circulation. Jpn Heart J 2003;44:601-612.
Mikkelsen L, Sheykhzade M, Jensen KA, Saber AT, Jacobsen NR, Vogel U, Wallin H, Loft
S, Møller P. Modest effect on plaque progression and vasodilatory function in
atherosclerosis-prone mice exposed to nanosized TiO(2). Part Fibre Toxicol 2011;8:32.
51
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
Morimoto Y, Hirohashi M, Ogami A, Oyabu T, Myojo T, Todoroki M, Yamamoto M,
Hashiba M, Mizuguchi Y, Lee BW, Kuroda E, Shimada M, Wang WN, Yamamoto K,
Fujita K, Endoh S, Uchida K, Kobayashi N, Mizuno K, Inada M, Tao H, Nakazato T,
Nakanishi J, Tanaka I. Pulmonary toxicity of well-dispersed multi-wall carbon nanotubes
following inhalation and intratracheal instillation. Nanotoxicology 2012;6:587-599.
Muhle H, Bellmann B, Creutzenberg O, Dasenbrock C, Ernst H, Kilpper R, MacKenzie
JC, Morrow P, Mohr U, Takenaka S. Pulmonary response to toner upon chronic
inhalation exposure in rats. Fundam Appl Toxicol 1991;17:280-299.
Nakanishi J, Gamo M. Risk Assessment of Manufactured Nanomaterials Titanium
dioxide (TiO2). Final report issued on July 22, 2011. NEDO project P06041 "Research and
development of nanoparticle characterization methods". NEDO, 2011.
Nakashima Y, Plump AS, Raines EW, Breslow JL, Ross R. ApoE-deficient mice develop
lesions of all phases of atherosclerosis throughout the arterial tree. Arterioscler Thromb
1994;14:133-140.
NIOSH. Occcupational exposure to Titanium dioxide. Current Intelligence Bulletin 63.
Cincinnati, OH: Department of Health and Human Services; Centers for Disease Control
and Prevention; National Institute for Occupational Safety and Health, 2011.
Noel A, Maghni K, Cloutier Y, Dion C, Wilkinson KJ, Halle S, Tardif R, Truchon G.
Effects of inhaled nano-TiO2 aerosols showing two distinct agglomeration states on rat
lungs. Toxicol Lett 2012;214:109-119.
Nurkiewicz TR, Porter DW, Hubbs AF, Stone S, Chen BT, Frazer DG, Boegehold MA,
Castranova V. Pulmonary nanoparticle exposure disrupts systemic microvascular nitric
oxide signaling. Toxicol Sci 2009;110:191-203.
Oberdorster G, Oberdorster E, Oberdorster J. Nanotoxicology: An emerging discipline
evolving from studies of ultrafine particles. Environ Health Perspect 2005;113:823-839.
OECD Environment HaSP. Titanium dioxide: Summary of the dossier. Series on the
Safety of Manufactured Nanomaterials: 73. Paris: OECD, 2016.
Pelclova D, Zdimal V, Fenclova Z, Vlckova S, Turci F, Corazzari I, Kacer P, Schwarz J,
Zikova N, Makes O, Syslova K, Komarc M, Belacek J, Navratil T, Machajova M,
Zakharov S. Markers of oxidative damage of nucleic acids and proteins among workers
exposed to TiO2 (nano) particles. Occup Environ Med 2016a;73:110-118.
Pelclova D, Zdimal V, Kacer P, Fenclova Z, Vlckova S, Komarc M, Navratil T, Schwarz J,
Zikova N, Makes O, Syslova K, Belacek J, Zakharov S. Leukotrienes in exhaled breath
condensate and fractional exhaled nitric oxide in workers exposed to TiO2 nanoparticles.
J Breath Res 2016b;10:036004.
52
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
Pelclova D, Zdimal V, Kacer P, Zikova N, Komarc M, Fenclova Z, Vlckova S, Schwarz J,
Makes O, Syslova K, Navratil T, Turci F, Corazzari I, Zakharov S, Bello D. Markers of
lipid oxidative damage in the exhaled breath condensate of nano TiO2 production
workers. Nanotoxicology 2017;11:52-63.
Pepys MB, Hirschfield GM. C-reactive protein: a critical update. J Clin Invest
2003;111:1805-1812.
Porter DW, Hubbs AF, Chen BT, McKinney W, Mercer RR, Wolfarth MG, Battelli L, Wu
N, Sriram K, Leonard S, Andrew M, Willard P, Tsuruoka S, Endo M, Tsukada T,
Munekane F, Frazer DG, Castranova V. Acute pulmonary dose-responses to inhaled
multi-walled carbon nanotubes. Nanotoxicology 2013;7:1179-1194.
Poulsen SS, Saber AT, Mortensen A, Szarek J, Wu D, Williams A, Andersen O, Jacobsen
NR, Yauk CL, Wallin H, Halappanavar S, Vogel U. Changes in cholesterol homeostasis
and acute phase response link pulmonary exposure to multi-walled carbon nanotubes to
risk of cardiovascular disease. Toxicol Appl Pharmacol 2015a;283:210-222.
Poulsen SS, Saber AT, Williams A, Andersen O, Købler C, Atluri R, Pozzebon ME,
Mucelli SP, Simion M, Rickerby D, Mortensen A, Jackson P, Kyjovska ZO, Mílhave K,
Jacobsen NR, Jensen KA, Yauk CL, Wallin H, Halappanavar S, Vogel U. MWCNTs of
different physicochemical properties cause similar inflammatory responses, but
differences in transcriptional and histological markers of fibrosis in mouse lungs. Toxicol
Appl Pharmacol 2015b;284.:16-32.
RAC. Titanium dioxide proposed to be classified as suspected of causing cancer when
inhaled. ECHA's Committee for Risk Assessment (RAC). RAC, 2017.
Ramanakumar AV, Parent ME, Latreille B, Siemiatycki J. Risk of lung cancer following
exposure to carbon black, titanium dioxide and talc: Results from two case-control
studies in Montreal. Int J Cancer 2008;122:183-189.
Ridker PM, Hennekens CH, Buring JE, Rifai N. C-reactive protein and other markers of
inflammation in the prediction of cardiovascular disease in women. N Engl J Med
2000;342:836-843.
Rossi EM, Pylkkanen L, Koivisto AJ, Nykasenoja H, Wolff H, Savolainen K, Alenius H.
Inhalation exposure to nanosized and fine TiO2 particles inhibits features of allergic
asthma in a murine model. Part Fibre Toxicol 2010a;7:35.
Rossi EM, Pylkkänen L, Koivisto AJ, Vippola M, Jensen KA, Miettinen M, Sirola K,
Nykäsenoja H, Karisola P, Stjernvall T, Vanhala E, Kiilunen M, Pasanen P, Mäkinen M,
Hämeri K, Joutsensaari J, Tuomi T, Jokiniemi J, Wolff H, Savolainen K, Matikainen S,
Alenius H. Airway exposure to silica-coated TiO2 nanoparticles induces pulmonary
neutrophilia in mice. Toxicol Sci 2010b;113:422-433.
53
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
Saber AT, Jacobsen NR, Jackson P, Poulsen SS, Kyjovska ZO, Halappanavar S, Yauk CL,
Wallin H, Vogel U. Particle-induced pulmonary acute phase response may be the causal
link between particle inhalation and cardiovascular disease.
Wiley Interdiscip Rev Nanomed Nanobiotechnol 2014;6:517-531.
Saber AT, Jacobsen NR, Mortensen A, Szarek J, Jackson P, Madsen AM, Jensen KA,
Koponen IK, Brunborg G, Gutzkow KB, Vogel U, Wallin H. Nanotitanium dioxide
toxicity in mouse lung is reduced in sanding dust from paint. Part Fibre Toxicol
2012a;9:4.
Saber AT, Jensen KA, Jacobsen NR, Birkedal R, Mikkelsen L, Møller P, Loft S, Wallin H,
Vogel U. Inflammatory and genotoxic effects of nanoparticles designed for inclusion in
paints and lacquers. Nanotoxicology 2012b;6:453-471.
Saber AT, Lamson JS, Jacobsen NR, Ravn-Haren G, Hougaard KS, Nyendi AN, Wahlberg
P, Madsen AM, Jackson P, Wallin H, Vogel U. Particle-induced pulmonary acute phase
response correlates with neutrophil influx linking inhaled particles and cardiovascular
risk. PLoS One 2013;8:e69020.
SCHER/SCCP/SCENIHR. Risk assessment methodologies and approaches for genotoxic
and carcinogenic substances. European Commission Health & Consumer Protection,
2009. http://ec.europa.eu/health/ph_risk/committees/04_scher/docs/scher_o_113.pdf
Schmid O, Cassee FR. On the pivotal role of dose for particle toxicology and risk
assessment: Exposure is a poor surrogate for delivered dose. Part Fibre Toxicol
2017;14:52.
Scientific Committee on Consumer Safety (SCCS). Opinion on Titanium Dioxide (nano
form) as UV-filter in sprays. SCCS/1583/17. Luxembourg: European Commission, 2017.
Shi H, Magaye R, Castranova V, Zhao J. Titanium dioxide nanoparticles: a review of
current toxicological data. Part Fibre Toxicol 2013;10:15.
Stapleton PA, Hathaway QA, Nichols CE, Abukabda AB, Pinti MV, Shepherd DL,
McBride CR, Yi J, Castranova VC, Hollander JM, Nurkiewicz TR. Maternal engineered
nanomaterial inhalation during gestation alters the fetal transcriptome.
Part Fibre Toxicol 2018a;15:3.
Stapleton PA, McBride CR, Yi J, Abukabda AB, Nurkiewicz TR. Estrous cycle-dependent
modulation of in vivo microvascular dysfunction after nanomaterial inhalation.
Reprod Toxicol 2018b;78:20-28.
Steinmetz A, Hocke G, Saile R, Puchois P, Fruchart JC. Influence of serum amyloid A on
cholesterol esterification in human plasma. Biochim Biophys Acta 1989;1006:173-178.
Stockmann-Juvela T, Taxell P, Santonen T. Formulating occupational exposure limits
values (OELs) (inhalation & dermal). Scaffold SPD7. [European Community's Seventh
Framework Programme (FP7/20072013)]. Scaffold Public Documents, 2014.
54
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
Stone V, Miller MR, Clift MJD, Elder A, Mills NL, Møller P, Schins RPF, Vogel U,
Kreyling WG, Alstrup JK, Kuhlbusch TAJ, Schwarze PE, Hoet P, Pietroiusti A, De
Vizcaya-Ruiz A, Baeza-Squiban A, Teixeira JP, Tran CL, Cassee FR. Nanomaterials
versus ambient ultrafine particles: An opportunity to exchange toxicology knowledge.
Environ Health Perspect 2017;125:106002.
Tran CL, Cullen RT, Buchanan D, Jones AD, Miller BG, Searl A, Davis JMG, Donaldson
K. Investigation and prediction of pulmonary responses to dust Part II. In: Investigations
into the pulmonary effects of low toxicity dusts. Contract Research Report: 216. Suffolk,
UK: HSE Books, 1999.
Virmani R, Kolodgie FD, Burke AP, Finn AV, Gold HK, Tulenko TN, Wrenn SP, Narula
J. Atherosclerotic plaque progression and vulnerability to rupture: Angiogenesis as a
source of intraplaque hemorrhage. Arterioscler Thromb Vasc Biol 2005;25:2054-2061.
Warheit DB, Frame SR. Characterization and reclassification of titanium dioxide-related
pulmonary lesions. J Occup Environ Med 2006;48:1308-1313.
Whitehead AS, Zahedi K, Rits M, Mortensen RF, Lelias JM. Mouse C-reactive protein.
Generation of cDNA clones, structural analysis, and induction of mRNA during
inflammation. Biochem J 1990;266:283-290.
World Health Organization. Cardiovascular Diseases (CVDs). World Health
Organization, 2018.
http://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds)
Xu H, Zhao L, Chen Z, Zhou J, Tang S, Kong F, Li X, Yan L, Zhang J, Jia G. Exposure
assessment of workplace manufacturing titanium dioxide particles. J Nanopart Res
2016;18.
Zhao L, Zhu Y, Chen Z, Xu H, Zhou J, Tang S, Xu Z, Kong F, Li X, Zhang Y, Li X, Zhang
J, Jia G. Cardiopulmonary effects induced by occupational exposure to titanium dioxide
nanoparticles. Nanotoxicology 2018;12:169-184.
55
BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren BEU, Alm.del - 2019-20 - Bilag 101: Orientering om NFA’s forslag til grænseværdier for fem kemiske stoffer, fra beskæftigelsesministeren
2130137_0060.png
Lersø Parkallé 105
DK-2100 Copenhagen
T +45 3916 5200
F +45 3916 5201
E [email protected]
W www.nfa.dk