Using adverse outcome pathway analysis to identify gaps in high ...

18
Using adverse outcome pathway analysis to identify gaps in high-throughput screening for thyroid disruption [email protected] August 2014 Katie Paul Disclaimer: The views herein are those of the speaker and do not represent Bayer CropScience or the US Environmental Protection Agency.

Transcript of Using adverse outcome pathway analysis to identify gaps in high ...

Page 1: Using adverse outcome pathway analysis to identify gaps in high ...

Using adverse outcome pathway analysis to identify gaps in high-throughput screening for thyroid disruption [email protected]

August 2014 Katie Paul Disclaimer: The views herein are those of the speaker and do not represent Bayer CropScience or the US Environmental Protection Agency.

Page 2: Using adverse outcome pathway analysis to identify gaps in high ...

Objectives A view of the importance of thyroid hormone to neurodevelopment in rodents and humans.

An adverse outcome pathway for thyroid hormone perturbation.

A comparison of available rodent thyroid data in ToxRefDB versus the available high-throughput screening assay data in ToxCast.

Opportunities for assay development.

Current barriers to the use of AOPs in chemical screening.

Page 3: Using adverse outcome pathway analysis to identify gaps in high ...

Maternal thyroid hormones modulate fetal brain development; thus screening for developmental Δ[TH] may identify neurotoxicants.

Maternal thyroid hormone (free T4)

0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 30 32 34 36 38 40 Human Gestation Week

Thyroid development

Pituitary development

Hypothalamus development

Feta

l thy

roid

re

gula

tion

Brain stem

Cerebellum

Midbrain

Hippocampus and substructures

Pallium and Cortical structures

Olfactory structures

Hypothalamus and thalamus

Synaptogenesis

Myelinogenesis

Gliogenesis

Feta

l Neu

roge

nesi

s

Neural tube

Fetal TSH

Thyroid hormone production

Thyroid receptors

Adapted from Howdeshell (2002) EHP.

Page 4: Using adverse outcome pathway analysis to identify gaps in high ...

Moderate maternal thyroid hormone disruption

Cognitive

and brain abnormalities.

(Barone et al., 2000; Berbel et al. 2009; Cuevas et al., 2005; Howdeshell, 2002; Morreale de Escobar et al., 2000; Rice et al., 2000; Zoeller et al., 2000)

Human maternal hypothyroxinemia in the

first trimester

• ↓ behavioral response @ 3 wks (Kooistra et al. 2006); • ↓ psychomotor development @ 10, 12, 24 mos (Pop et al., 1999; Pop et al., 2003); • ↓ motor coordination/socialization at 18 mos (Berbel et al. 2009); • Language delays at 18 and 30 mos (Henrichs et al. 2010); • Small IQ point ↓ at 7-9 yrs (Haddow et al. 1999).

Rat models of maternal T4

insufficiency

• ∆ cytoarchitecture (Auso et al., 2004; Cuevas et al., 2005; Lavado-Autric et al., 2003; Sharlin et al. 2008);

• ∆ synaptic calcium regulation and myelination (Ibarrola et al., 1997; Iniguez et al., 1996); • Gene expression ∆ :synaptic calcium/transmission, myelination, and developmental cell adhesion (Morreale de Escobar et al., 2008; Morreale de Escobar et al., 2000; Morreale de Escobar et al., 2004, Royland et al.,2008).

Page 5: Using adverse outcome pathway analysis to identify gaps in high ...

AOPs provide a framework to illustrate existing understanding of how an initial chemical action is potentially associated with apical outcomes.

Reproduced from Ankley et al. (2010) ETC

Page 6: Using adverse outcome pathway analysis to identify gaps in high ...

AOP analysis, high-throughput screening, and informing predictive models iteratively

Use AOPs to propose HTS

assays for MIEs or key events

Validate HTS assays for purpose

(relevance and reliability)

Compare to existing in vivo and in vitro data

Test assays

Reference Chemicals

Chemical library

Determine the utility of the

assay/assay set HTS data or

in vivo repository,

eg ToxRefDB

Reproduced from Reif et al. (2010) EHP Cycle of assay and assay set development to generate the best available information for predictive models and prioritization (Adapted from EPA Factsheet, Monica Linnenbrink)

Page 7: Using adverse outcome pathway analysis to identify gaps in high ...

The TH system is regulated at several nodes, each corresponding to a potential MIE.

UGTs, SULTs, Transport

TPO

TBG/TTR/Alb

Peripheral deiodinases

TRsubtype

NIS

T4, T3

Propylthiouracil, methimazole, Benzophenone-2, isoflavones, etc.

Perchlorate, bromate, some small organics

PCBs, PTU, FD&C Red No3 dye, etc.

?

Figure reproduced from Reif et al. 2010 EHP

ToxPi predictive tool for endocrine disruptors

Several MIEs do not correspond to a medium- or high-throughput screening assay in ToxCast/Tox21.

Page 8: Using adverse outcome pathway analysis to identify gaps in high ...

Thyroperoxidase (TPO) inhibition

Na+I- symporter (NIS) inhibition

Xenobiotic nuclear receptor activation

Deiodinase inhibition

Serum binding interference

Thyroid receptor (TR) binding

↓ TH synthesis

↑ Phase II catabolism

↑ Hepatic transport

↓ serum T4/T3 ↓ tissue [TH] concentrations

Altered neurodevelopment

Altered metamorphosis

(amphibian)

Rodent thyroid histopathology and

weight ↑

↓ Tissue T4T3 conversion

Molecular-initiating events Key events Adverse outcomes

Developed with Kevin Crofton, US EPA; Murk et al, 2013 Toxicol In Vitro; Crofton 2008 Int J Androl.

Thyroid hormone transport

interference

Multiple molecular-initiating events converge on common key events to proceed to adverse outcomes.

Page 9: Using adverse outcome pathway analysis to identify gaps in high ...

Altered neurodevelopment

Altered metamorphosis

(amphibian)

Rodent thyroid histopathology and

weight ↑

Adverse outcomes

Ecological concern

Conserved across

species?

Not relevant to human

health

Rodent thyroid tumors resultant to TSH stimulation are not relevant to human carcinogenesis.

Parameter Rodent Human Sources

Differentiated tumor types

Follicular Follicular (20%) or papillary (60%)

1Williams (1995) Mechanisms and pathogenesis of thyroid cancer in animals and man. Mutation Research; 333: 123-129. 2McClain (1989) The Significance of Hepatic Microsomal Enzyme Induction and Altered Thyroid Function inRats: Implications for Thyroid Gland Neoplasia. Toxicologic Pathology; 17(2): 294-206.

3Hill et al. (1998) Risk assessment of Thyroid Follicular Cell Tumors. Environmental Health Perspectives; 106(8): 447-457. 4Hard (1998) Recent Developments in the Investigation of Thyroid Regulation and Thyroid Carcinogenesis. Research Reviews; 106(8): 427-436.

Etiology TSH stimulation (nonmutagenic) or mutagenic agents

Mutagenic agents (coupled with growth stimulation)

Serum TSH 6-60X 1X

• Tumors as a biomarker: tumors themselves are not relevant to humans, but they may indicate the potential for thyroid hormone perturbation across species.

• Limitations of the biomarker: Changes in T4 and/or T3, particularly transient changes or changes only

in T4, may not result in a rodent thyroid tumor.

Page 10: Using adverse outcome pathway analysis to identify gaps in high ...

Rodent thyroid tumors may be a biomarker for potential Δ[TH].

Phenobarbital example: rodent

Indirect CAR activation

↑ Phase II catabolism

↑ Hepatic transport

↓ serum T4/T3 ↑TSH

↓ tissue [TH] concentrations

Altered neurodevelopment

Rodent thyroid histopathology and

weight ↑

Vehi

cle

50 m

g/kg

phe

noba

rbita

l, b.

i.d.,

5 w

ks.

h

h

Reproduced from Smith PJ et al. (1991) Tox Path.

Moderaypertro

and yperpla

te phy

sia

Many rodent studies available to support neurological changes, including: ↓brain weight, cerebellum weight, Purkinje and granular cells in the cerebellum, granule cells in the hippocampus, performance in motor activity/learning tests (reviewed by Slikker and Chang, Handbook of Developmental Toxicology and Costa et al. 2005 Ann Rev Pharm Tox)

Page 11: Using adverse outcome pathway analysis to identify gaps in high ...

Rodent thyroid tumors may be a biomarker for potential Δ[TH].

Phenobarbital example: moderate human plausibility

Indirect CAR activation and/or

PXR at high doses

↑ Phase II catabolism

↑ Hepatic transport

↓ serum T4/T3 ↓ tissue [TH] concentrations

Altered neurodevelopment

Rotroff et al. 2010 JTEH Part B;

Paul et al. 2013 Tox In Vitro;

many others

In vitro: Rotroff et al. 2010 JTEH Part B; Ritter et al. 1999

Hepatology (UGT1A1 ↑ at 10x

Tx serum conc [Strolin et al 2005]); in vivo evidence is

lacking

Generally only in specific cases or above therapeutic

concentrations (Strolin et al. 2005, Fun Clin Pharm)

Apparent data gap Mixed evidence; some supporting

studies suggest in utero exposure

results in ↓ verbal scores, IQ, spatial processing (Verotti et al. 2014 Repro

Tox).

Perhaps difficult to separate apical outcomes from potential GABA-mimetic or direct neurological effects.

Similar AOP for PCBs or other chemicals also have data gaps despite some human plausibility, the link to effects in humans is weak (Crofton & Zoeller 2005 Crit Rev in Toxicol; Zoeller & Crofton 2005 Crit Rev in Toxicol).

Page 12: Using adverse outcome pathway analysis to identify gaps in high ...

Anchoring HTS assay data from ToxCast to ToxRefDB thyroid endpoints could illustrate how well current screening efforts capture potential thyroid disruption.

*239 chemicals with Phase I, Phase II, and ToxRefDB data

Study type Species Endpoints Subchronic Rat, Dog Thyroid gland weight, nonproliferative

pathology, proliferative pathology, thyroid gland effect (e.g., gross change)

Chronic Rat, Mouse

Thyroid gland weight, nonproliferative pathology, proliferative pathology, thyroid gland effect (gross change)

Multi-generation Rat Nonproliferative pathology, thyroid gland weight, thyroid tumor

Page 13: Using adverse outcome pathway analysis to identify gaps in high ...

A binary view of thyroid changes in ToxRefDB versus in vitro bioactivity prediction.

ToxRefDB (+)

ToxRefDB (-)

ToxCast (+)

True Positive

False Positive

(?)

ToxCast (-)

False Negative

True Negative

(?)

Lack of concordance between in vivo thyroid

effects and current assay battery underscores that

important MIEs are missing from the assay battery.

Page 14: Using adverse outcome pathway analysis to identify gaps in high ...

Missing MIEs from the current screening battery for ToxCast/Tox21.

Thyroperoxidase (TPO) inhibition

Na+I- symporter (NIS) inhibition

Xenobiotic nuclear receptor activation

Deiodinase inhibition

Serum binding interference

Thyroid receptor (TR) binding

↓ TH synthesis

↑ Phase II catabolism

↑ Hepatic transport

↓ serum T4/T3 ↓ tissue [TH] concentrations

Altered neurodevelopment

Altered metamorphosis

(amphibian)

Rodent thyroid histopathology and

weight ↑

↓ Tissue T4T3 conversion

Molecular-initiating events Key events Adverse outcomes

Developed with Kevin Crofton, US EPA; Murk et al, 2013 Toxicol In Vitro; Crofton 2008 Int J Androl.

Thyroid hormone transport

interference

Page 15: Using adverse outcome pathway analysis to identify gaps in high ...

Addition of assays for MIEs including TPO may improve prediction for rodent thyroid pathology and/or Δ[TH].

PTU/MMI: Human

evidence Inferred from

studies of hypothyroxinemia

Page 16: Using adverse outcome pathway analysis to identify gaps in high ...

A hypothetical schematic of how an initial screening battery could relate to confirmatory models.

Page 17: Using adverse outcome pathway analysis to identify gaps in high ...

Current barriers to the use of thyroid AOPs in a screening and prioritization context

Barrier Action merited

Internationally-accepted AOPs for thyroid disruption with clear delineation of data gaps.

Work-in-progress with the OECD and the AOP-Wiki to establish AOPs for thyroperoxidase inhibition and xenobiotic catabolism; more effort should be applied to cover all MIEs (Murk et al., 2013).

Not enough MIEs from thyroid AOP are covered by assay approaches.

Increased screening activity, increased assay development, investment of resources to run lower throughput screening models, increased development of QSAR models for use in screening for particular MIEs rather than for apical thyroid outcomes.

Lack of quantitative linkages between MIE and key events in thyroid AOPs.

Quantitative evaluation of key events in thyroid perturbation in in vitro and in vivo models.

Lack of defined targeted, confirmatory models, and a decision-tree for how HTS assay relate to these models in a workflow.

Quantitative and qualitative assessment of how models relate; development and characterization of more complex medium-throughput models (in vitro and in vivo).

Lack of definitive evidence of human concordance for apical outcomes.

Plausibility of interrelated key events should be assessed in models, but mechanistic human data on the concordance of apical outcomes will likely not be available.

Ideal outcome: Combine AOP-based hazard prediction with exposure prediction to prioritize chemicals for any further evaluation in models for greater confidence.

Page 18: Using adverse outcome pathway analysis to identify gaps in high ...

Thank you! Especially colleagues at Bayer CropScience, Human Safety Regulatory Toxicology and Kevin Crofton, US EPA

Katie Paul August 2014