using human data to protect the public's health

23
Regulatory Toxicology and Pharmacology 33, 234–256 (2001) doi:10.1006/rtph.2001.1469, available online at http://www.idealibrary.com on Using Human Data to Protect the Public’s Health Michael L. Dourson, *,1 Melvin E. Andersen,Linda S. Erdreich,and Judith A. MacGregor§ * Toxicology Excellence for Risk Assessment, Cincinnati, Ohio 45223; Colorado State University; Exponent; and §Toxicology Consulting Services Received February 26, 2001 The value of using human data in the assessment and management of risk is evaluated. Although the use of such data has a long and successful history with environmental contaminants and the development of drugs and commercial chemicals, recent deliberations within the Environmental Protection Agency (EPA) have questioned this practice in part. Specifically, we evaluate the degree to which reference doses (RfDs) and reference concentrations (RfCs) derived from human data on EPA’s Integrated Risk Information System (IRIS) differ with RfDs and RfCs that we es- timate from experimental animal data. We also use several minimal risk levels of the Agency for Toxic Sub- stances and Disease Registry (ATSDR) and tolerable intakes of Health Canada in this comparison. Human- based RfDs are more than threefold lower than the corresponding animal-based RfDs for 23% of the com- parisons. Human- based RfDs or RfCs are lower than corresponding animal-based RfDs or RfCs for 36% of the comparisons. Furthermore, for 10 of 43 possible comparisons, insufficient experimental animal data are readily available or data are inappropriate to es- timate either RfDs or RfCs. We also discuss human pharmacokinetic data from volunteer studies and mechanistic studies with human tissues in vitro and demonstrate through a series of case discussions that utilization of such data is important when making de- cisions to protect exposed individuals. Moreover, phys- iologically based pharmacokinetic (PBPK) modeling evaluates critical information in assessing interindi- vidual variability and identifying at-risk populations. Within the limits of our analysis, we conclude that the direct use and interpretation of human data, in con- junction with data gathered from experimental ani- mals, are public health protective policies that should be encouraged. C 2001 Academic Press Key Words: dose response; human; reference dose; reference concentration; risk; pharmacokinetics; pharmacodynamics; uncertainty factor. 1 To whom correspondence and reprint requests should be addressed at Toxicology Excellence for Risk Assessment, 1757 Chase Avenue, Cincinnati, OH 45223. Fax: 513-542-7487. E-mail: [email protected]. INTRODUCTION Is information from human studies the best way to judge the potential public health risk from chemicals in our environment? Should public health agencies strive to resolve the ethical questions and identify criteria to foster the conduct of human studies that might lead to better protection of the public’s health? Should scien- tists ignore available human data that might suggest a lower or higher risk value? What is a risk value? These issues have been a subject of much recent dis- cussion (e.g., EPA, 2000a; Russo, 2000; SOT, 2000) and a topic of increased attention among both scientists and policy makers. Answers to these questions are compli- cated and not necessarily straightforward. Perhaps the easiest question to answer is the last because it covers a narrower part of the NAS (1983, 1994) risk assess- ment and risk management paradigm—that of hazard identification and dose–response assessment. Conceiv- ably it should be the first question answered, since its response lays the foundation for the other questions in- volving more than the underlying science. We attempt to answer this question directly below. Other questions are discussed later. What Is a Risk Value? In its simplest form, a risk value is a given point on the dose–response curve associated with some probabil- ity of an outcome. In many cases, risk values are cho- sen to be associated with a level of zero or very small risk, usually referred to as “no appreciable risk.” A com- mon example of a risk value is a reference dose (RfD). A RfD is a point on the dose–response curve for a chem- ical of interest that is believed to be in the region of no adverse effect and is often used by risk managers to distinguish between the region of no adverse effect and the region of adverse effect. With either interpre- tation, however, the RfD does not distinguish between the region of any effect and that of no effect, because scientists often distinguish between a chemical’s abil- ity to cause an adverse effect and its ability to cause ef- fects that are not considered adverse. For example, risk assessors often distinguish among adaptive, adverse, 234 0273-2300/01 $35.00 Copyright C 2001 by Academic Press All rights of reproduction in any form reserved.

Upload: independent

Post on 10-Apr-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

Regulatory Toxicology and Pharmacology 33, 234–256 (2001)doi:10.1006/rtph.2001.1469, available online at http://www.idealibrary.com on

Using Human Data to Protect the Public’s Health

Michael L. Dourson,∗,1 Melvin E. Andersen,† Linda S. Erdreich,‡ and Judith A. MacGregor§∗Toxicology Excellence for Risk Assessment, Cincinnati, Ohio 45223; †Colorado State University; ‡Exponent;

and §Toxicology Consulting Services

Received February 26, 2001

The value of using human data in the assessmentand management of risk is evaluated. Although the useof such data has a long and successful history withenvironmental contaminants and the development ofdrugs and commercial chemicals, recent deliberationswithin the Environmental Protection Agency (EPA)have questioned this practice in part. Specifically, weevaluate the degree to which reference doses (RfDs)and reference concentrations (RfCs) derived fromhuman data on EPA’s Integrated Risk InformationSystem (IRIS) differ with RfDs and RfCs that we es-timate from experimental animal data. We also useseveral minimal risk levels of the Agency for Toxic Sub-stances and Disease Registry (ATSDR) and tolerableintakes of Health Canada in this comparison. Human-based RfDs are more than threefold lower than thecorresponding animal-based RfDs for 23% of the com-parisons. Human- based RfDs or RfCs are lower thancorresponding animal-based RfDs or RfCs for 36% ofthe comparisons. Furthermore, for 10 of 43 possiblecomparisons, insufficient experimental animal dataare readily available or data are inappropriate to es-timate either RfDs or RfCs. We also discuss humanpharmacokinetic data from volunteer studies andmechanistic studies with human tissues in vitro anddemonstrate through a series of case discussions thatutilization of such data is important when making de-cisions to protect exposed individuals. Moreover, phys-iologically based pharmacokinetic (PBPK) modelingevaluates critical information in assessing interindi-vidual variability and identifying at-risk populations.Within the limits of our analysis, we conclude that thedirect use and interpretation of human data, in con-junction with data gathered from experimental ani-mals, are public health protective policies that shouldbe encouraged. C© 2001 Academic Press

Key Words: dose response; human; reference dose;reference concentration; risk; pharmacokinetics;pharmacodynamics; uncertainty factor.

1 To whom correspondence and reprint requests should beaddressed at Toxicology Excellence for Risk Assessment, 1757Chase Avenue, Cincinnati, OH 45223. Fax: 513-542-7487. E-mail:[email protected].

INTRODUCTION

230273-2300/01 $35.00Copyright C© 2001 by Academic PressAll rights of reproduction in any form reserved.

Is information from human studies the best way tojudge the potential public health risk from chemicals inour environment? Should public health agencies striveto resolve the ethical questions and identify criteria tofoster the conduct of human studies that might lead tobetter protection of the public’s health? Should scien-tists ignore available human data that might suggest alower or higher risk value? What is a risk value?

These issues have been a subject of much recent dis-cussion (e.g., EPA, 2000a; Russo, 2000; SOT, 2000) anda topic of increased attention among both scientists andpolicy makers. Answers to these questions are compli-cated and not necessarily straightforward. Perhaps theeasiest question to answer is the last because it coversa narrower part of the NAS (1983, 1994) risk assess-ment and risk management paradigm—that of hazardidentification and dose–response assessment. Conceiv-ably it should be the first question answered, since itsresponse lays the foundation for the other questions in-volving more than the underlying science. We attemptto answer this question directly below. Other questionsare discussed later.

What Is a Risk Value?

In its simplest form, a risk value is a given point onthe dose–response curve associated with some probabil-ity of an outcome. In many cases, risk values are cho-sen to be associated with a level of zero or very smallrisk, usually referred to as “no appreciable risk.” A com-mon example of a risk value is a reference dose (RfD).A RfD is a point on the dose–response curve for a chem-ical of interest that is believed to be in the region ofno adverse effect and is often used by risk managersto distinguish between the region of no adverse effectand the region of adverse effect. With either interpre-tation, however, the RfD does not distinguish betweenthe region of any effect and that of no effect, becausescientists often distinguish between a chemical’s abil-ity to cause an adverse effect and its ability to cause ef-fects that are not considered adverse. For example, riskassessors often distinguish among adaptive, adverse,

4

USING HUMAN DATA TO PROTECT THE PUBLIC’S HEALTH 235

compensatory, and critical effects.2 In this process theconcepts of severity of effect, homeostasis, and hormesisare often discussed.

Specifically, a RfD is defined as “an estimate (withuncertainty spanning perhaps an order of magnitude)of a daily exposure to the human population (includ-ing sensitive subgroups) that is likely to be withoutan appreciable risk of deleterious effects during a life-time” (Barnes and Dourson, 1988). The reference con-centration (RfC), where dose is expressed as a concen-tration, is similarly defined and applies to inhalationexposures (Jarabek, 1994, 1995). A measure or esti-mate of human exposure is often integrated with arisk value such as a RfD or RfC to develop a simplerisk characterization, such as a maximum contaminantlevel goal (MCLG) in drinking water. This MCLG isthen considered with other information in developinga risk management position, such as a maximum con-taminant level (MCL). This MCL is enforceable and isdesigned to protect the public’s health. EPA developsother risk characterizations and risk management de-cisions for other media. Such characterizations mightinvolve more complex dose–response assessments andexposure assessments. Health organizations aroundthe world use risk values and their associated ex-posure measures in similar fashion when conductingrisk characterizations and arriving at risk managementdecisions.

In some cases, scientists postulate that a chemicalmay not have a distinguishable region of adverse ef-fect and no adverse effect in the dose–response curve.In such cases, the adverse effect might occur at anydose, with more and more individuals being affected atgreater doses (such as what might occur with cancer), orthe adverse effect will not occur at any dose. With manychemicals this distinction is debated, because the scien-

tific data are insufficient to provide certainty in conclu-

2 Below are common definitions of types of effects. These effects arenot always clear-cut categories, but often represent a continuum. Dif-ferent individuals are expected to have different capacities for adap-tive and compensatory effects.

• Adaptive effects are not considered to be adverse. They enhancean organism’s performance as a whole and/or its ability to withstanda challenge. An increase in hepatic smooth endoplasmic reticulum isan example of an adaptive effect, if hepatic metabolism reduces thechemical’s toxicity.• Adverse effects are biochemical changes, functional impair-

ments, or pathologic lesions that affect the performance of the wholeorganism or reduce an organism’s ability to respond to additional en-vironmental challenges (EPA, 2000b).• Compensatory effects are not considered to be adverse. They

maintain overall function without enhancement or significant cost.Increased respiration due to metabolic acidosis is an example of acompensatory effect.• Critical effects are the first adverse effects or their known pre-

cursors that occur as dose rate increases (EPA, 2000b).• Severity is the degree to which an effect changes and impairs the

functional capacity of an organ system.

sions in the area of the dose–response curve used forinference.

How Are Human Data Used?

Human data can be used in several ways in a haz-ard identification and dose–response assessment. Forhazard identification, human data can be used alone toevaluate a finding or it may be used in concert with ex-perimental animal data to provide weight-of-evidencethat an observed association between exposure and re-sponse is actually caused by the exposure. In general,when used alone, human epidemiological data are in-sufficient evidence to strongly support causality (thereare notable exceptions to this rule such as arsenic). Inthe case of human data associated with clinical stud-ies or clinical interventions (such as for tamoxifen), thestrength of the causal association is higher than withenvironmental epidemiological data. The main reasonsfor this difference pertain to limitations in the mea-surement of exposures in environmental epidemiologi-cal studies and the inability to control or include con-founding variables that could also be associated withthe observed toxicity.

In dose–response assessment, human data regard-ing the no-observed-adverse-effect level (NOAEL) canbe used directly as the basis of a RfD or RfC. The pres-ence of human data obviates the necessity of extrapo-lating from animals to humans; therefore, human stud-ies, when available, are given first priority, with animaltoxicity studies serving to complement them (Barnesand Dourson, 1988). However, using human data in thisfashion requires that the human study be judged to beof at least comparable quality to an animal study thatmight be used to determine such a risk value. Moreover,the human study must conform to the highest stan-dards associated with the conduct and evaluation of sci-entific data and include informed consent. Human datathat are not directly useful as the basis of the NOAELvalue can also be compared with animal data to de-termine the most appropriate interspecies uncertaintyfactor, rather than a default factor of 10 (Dourson et al.,1996).

Human data can also be used as a guide to determinethe appropriate toxicological end point to be consideredfor use in the hazard identification. In this regard,human case studies and incident reporting systemsor other in vivo or in vitro experiments could bevaluable when coupled with the more quantitative andbetter-controlled experimental animal toxicity studies.Used in this fashion, the human data can be seen toadd mechanistic understanding of the critical effectdetermined from animals or to otherwise limit or boundthe likely estimates of risk determined from the animalwork in the dose–response assessment. They can alsoidentify human effects that are not detected in animal

studies, such as with the cardiac valvular defects

236 DOURSON ET AL.

associated with the use of the drugs for weight loss(FDA, 1997).

What Are the Types of Human Data?

To date, the human data that have been used in riskassessments include a wide variety of types, based pri-marily on the different ways in which health data onhumans can be obtained. Human data range from anec-dotal case reports to systematically designed epidemi-ological studies of an exposed cohort. Epidemiologicalstudies use several different approaches for collectingdata, including surveillance, public health statistics, ge-ographic correlation studies, and the cohort and case–control studies designed to identify associations andsometimes to support inferences about cause and effect.Human data also include experimental exposures of in-dividuals; these are typically short-term studies thatinvolve doses below a hypothesized threshold of adverseeffect.

Epidemiological studies are routinely based on in-advertent exposures that may occur in the workplace,by unusual natural contamination, or as the after-math of an accident such as an explosion or indus-trial release. However, deliberate exposure to humanshas been used for centuries to test therapeutic proce-dures (Lee, 1980). Human studies are routinely usedin drug development to evaluate the efficacy or safetyof a therapeutic or diagnostic regimen. Clinical stud-ies, or phase I clinical trials, are designed to identifya safe dose of a potential therapeutic or preventiveagent. These studies are an intrinsic part of medicine,and methods for the design and statistical analysis ofthese studies are discussed in most statistical textbooks(Elwood, 1998).

The human experimental studies used for agents inthe environment generally measure biochemical and/orphysiological changes related to the anticipated adverseresponse in order to obtain quantitative information,usually for comparison with animal data. Criteria forboth Phase I clinical trials and experimental studiesin humans of environmental exposures include volun-tary participation. Exposures to environmental chem-icals are intended to produce minimal or no immedi-ate adverse effects and no irreversible adverse effects(e.g., these studies usually examine an end point in theadaptive or compensatory response range). In the caseof environmental exposures, studies are designed formultiple reasons such as to aid in understanding howexposures might be measured in a general populationor to provide additional information for species com-parisons and reduce uncertainty. In both cases, thesestudies are aimed at improving health. In the case ofPhase I clinical trials for drugs, studies are designed toidentify dosing levels that can be tolerated without anyserious or unacceptable side effects and to serve as a

guide for dose selection for future studies.

Approximately 7 of 8 potential drugs fail duringclinical development and are subsequently not used(DeGeorge, 1999). For the 1 of 8 that are commerciallydeveloped, it is somewhat unlikely that a healthy vol-unteer would subsequently develop a need for the samepharmaceutical. In contrast, experimental studies forenvironmental contaminants have the potential for im-proving the risk estimates for chemicals for which thevolunteers and the rest of the general public may morelikely be exposed. Such improvement may have an indi-rect public health benefit. For example, the use of her-bicides and pesticides may indirectly benefit health ifthe agents reduce organisms that are disease vectors,or increase accessibility to foods needed for a healthydiet.

For evaluation in this paper all RfDs and RfCs basedon human data were selected from EPA’s IRIS (EPA,2000b). The human data that formed the basis of theseRfDs and RfCs were of all types, for example: case re-ports of argyria from exposure to silver, epidemiologicalstudies of populations exposed to natural arsenic or tomethyl mercury from environmental contamination, asurveillance epidemiology study for methemoglobine-mia from nitrate exposure, population studies of flu-orosis, and experimental studies of aldicarb, barium,warfarin, and zinc.

Purpose of This Research

The purpose of this research is to compare establishedRfDs and RfCs based on human data with those we esti-mate based on experimental animal data, and to showthe use of human toxicokinetic and toxicodynamic in-formation for estimating noncancer and cancer risk. Wealso briefly discuss the use and interpretation of humandata, in conjunction with data gathered from experi-mental animals, as a public health protective policy.

METHODS

Methods Used for Comparison of RfDs and RfCs

We chose to use the complete listing of noncancer riskvalues, that is RfDs and RfCs, based on human data asfound in EPA’s IRIS database (EPA, 2000b). RfDs andRfCs found in IRIS have gone through an extensive andrigorous development process, including internal peerreview and unanimous acceptance within EPA. IRIS isnot the only source of such information, of course (see forexample www.tera.org/iter), but we used IRIS becauseit is convenient, reasonably robust, objective and re-spected (although many of the risk values are outdated).We compared these human-based RfDs and RfCs withRfDs and RfCs we estimate from readily available ex-perimental animal data, mainly from IRIS, based onEPA dose–response assessment methods (Barnes andDourson, 1988; Dourson, 1994; Jarabek, 1994, 1995;

EPA, 1994). We chose not to do a similar comparison

USING HUMAN DATA TO PROTECT THE PUBLIC’S HEALTH 237

for cancer risk values because of the added complex-ity of developing risk values from experimental animaldata, although such work may be pursued in the future.

Our choice of appropriate experimental animal toxic-ity data to develop a RfD or RfC for comparison with anexisting human value depended primarily on the avail-ability of the experimental animal data. Confidence inthe resulting comparison depends in part on the confi-dence one has in the human-based RfD or RfC (see Ta-bles 1 and 2, which present confidence statements forrisk values on IRIS), and on the relevance of the resultsin animals to the critical effect shown in humans. In de-scending order of importance, we selected experimentalanimal data to match:

• The dose–response curve of the critical effect inthe human study [e.g., comparison of the dose–responsecurves of red blood cell cholinesterase (RBC) inhibition];or• The benchmark dose (BMD), no-observed-adverse-

effect level (NOAEL), or lowest-observed-adverse-effectlevel (LOAEL) of the critical effect in the humanstudy (e.g., comparison of RBC cholinesterase inhibi-tion NOAELs).

If matching data were not available, we then selec-ted:

• A BMD, NOAEL or LOAEL of a closely related ef-fect found in animals and compared it to the criticaleffect in humans (e.g., comparison of any clinical signsof cholinesterase inhibition); or• The most sensitive effect found in animals and com-

pared it to the critical effect in humans (e.g., comparisonof cholinesterase inhibition with liver toxicity).

We recognized that pharmacokinetics and pharma-codynamics (discussed later) may impact such compar-isons and conclusions drawn from these comparisons.Furthermore, the available experimental animal datamay not include a species relevant to humans, and anysuch comparisons of these animal data with the hu-man data may not be sufficiently predictive of effectsin humans. Therefore, these comparisons of establishedhuman-based RfDs and RfCs with those we estimatedfrom available animal data should be considered alongwith information on pharmacokinetics and pharmaco-dynamics in determining the usefulness of human data.

We developed animal-based RfDs and RfCs directlyfrom the existing information on EPA’s IRIS, and notfrom a thorough review of the original literature. Insome cases, EPA’s IRIS states an alternative RfD orRfC based on animal studies, and we used these alterna-tive values instead of estimating them. Occasionally, wecompared animal versus human risk values of ATSDR,EPA, and Health Canada as described on Toxicol-ogy Excellence for Risk Assessment’s (TERA) Interna-tional Toxicity Estimates for Risk (ITER) database (see

TERA, 2000). Please note that the animal-based RfDs

and RfCs that we derived have not undergone a rigor-ous peer review. Thus, the animal-based RfDs and RfCsthat we provide should only be considered as interim,subject to change with additional data and/or analysis.

In all cases, we developed the experimental animal-based RfDs and RfCs assuming no relevant human datawere available. Therefore, we used the default uncer-tainty factor of 10-fold (for RfDs) or 3-fold (for RfCs)for experimental animal to human extrapolation. Re-cent data and analysis by the EPA and others allow theuse of specific human and animal toxicity, toxicokineticand toxicodynamic data to affect the value of this andother uncertainty factors (Renwick, 1993; IPCS, 1994;Dourson, 1994; Dourson et al., 1996; see also discussionbelow).

In the development of the animal-based RfDs andRfCs, we used the same database uncertainty factor andmodifying factor as found in EPA’s IRIS with one ex-ception (the footnote for nitrite in Table 1 explains thisexception). This decision is reasonable because the useof these factors, and the choice of other potential factorssuch as that recommended under the Food Quality Pro-tection Act, reflects confidence in the overall database(EPA, 1999), which is the basis of both animal- andhuman-based RfDs and RfCs.

Methods for Pharmacokinetic Modeling

Making adjustments to animal results. Chemicalrisk assessments for cancer and noncancer end pointsare moving toward a common, harmonized methodology(Barton et al., 1998). The basic steps in the approachare the establishment of a point of departure that maybe a NOAEL, a LOAEL, or a BMD estimated from ex-perimental data as described above. In the inhalationreference concentration methodology (EPA, 1994), thepoint of departure value is adjusted for differences inexposure duration to account for the fact that animalsare exposed for less than 24 h per day while the expo-sure of interest for humans is continuous lifetime ex-posures. This duration-adjusted value is then correctedto take into account dosimetry differences expected be-tween animals and human in order to provide a humanequivalent concentration (HEC). These steps affect thenumerator in the RfC equation. Several uncertainty fac-tors are then applied to account for interspecies differ-ences between test animals and humans and interindi-vidual differences among humans. The HEC is dividedby these uncertainty factors. Human pharmacokineticand dosimetry data play potentially important roles inestimating the HEC and in assessing the magnitude ofthe uncertainty factors.

Mode of action and target tissue dose. The modeof action for a chemical entails the set of steps thatare involved in causing toxicity following exposure tothat particular chemical. Target tissue dose is the form

of the chemical, i.e., parent compound, metabolite, peak

238 DOURSON ET AL.

TA

BL

E1

Su

mm

ary

ofU

.S.E

PA

’sR

fDs

onIR

ISas

ofM

ay20

00B

ased

onH

um

anD

ata1

Ch

emic

aln

ame

NO

AE

L,

Un

cert

ain

tyfa

ctor

3R

fDra

tio:

(as

onE

PA

’sL

OA

EL

orR

fDH

um

anto

IRIS

)S

peci

e/T

ype

ofst

udy

BM

D2

Cri

tica

leff

ect(

s)T

otal

HA

LS

DM

FR

fDco

nfi

den

cean

imal

Ald

icar

bH

um

anex

peri

men

tal

0.01

Cli

nic

alsi

gns

ofbl

ood

or10

101

11

11

10−3

Med

ium

Eit

her

1ga

vage

plas

ma

chol

ines

tera

se(c

lin

ical

inh

ibit

ion

sign

s)or

Rat

deve

lopm

enta

l0.

125

Cli

nic

alsi

gns

ofch

olin

este

rase

100

1010

11

11

10−3

Med

ium

3(p

lasm

aga

vage

inh

ibit

ion

Ch

E)

Dog

52-w

eek

feed

ing

0.02

84P

lasm

ach

olin

este

rase

100

1010

11

11

10−4

Med

ium

inh

ibit

ion

Ars

enic

,H

um

anep

idem

iolo

gy0.

0008

Ski

nle

sion

san

dpo

ssib

le3

31

11

11

10−4

Med

ium

0.1

inor

gan

icdr

inki

ng

wat

erva

scu

lar

com

plic

atio

ns

Rat

13-w

eek

feed

ing5

2.6

(L)

Bla

dder

hyp

ertr

oph

yan

d10

0010

103

31

13×

10−3

Low

hyp

erpl

asia

Bar

ium

Hu

man

expe

rim

enta

l,0.

21In

crea

sed

bloo

dpr

essu

re3

31

11

11

10−2

Med

ium

0.4

epid

emio

logi

cal

drin

kin

gw

ater

Rat

chro

nic

drin

kin

g45

Incr

ease

dki

dney

wei

ght

300

1010

11

31

10−1

Med

ium

wat

erB

aygo

nH

um

anex

peri

men

tal

0.36

(L)

Mil

dch

olin

ergi

csy

mpt

oms,

100

101

101

11

10−3

Med

ium

0.8

sin

gle

dose

RB

Cch

olin

este

rase

inh

ibit

ion

Dog

12-m

onth

feed

ing

5(L

)C

hol

ines

tera

sein

hib

itio

n10

0010

1010

11

15×

10−3

Med

ium

Ben

zoic

acid

Hu

man

anec

dota

l4.

4N

oad

vers

eef

fect

sob

serv

ed1

11

11

11

4M

ediu

m10

06

diet

ary

expo

sure

Mou

sech

ron

ic40

(L)

Dec

reas

edre

sist

ance

tost

ress

1000

1010

101

11

10−2

Low

expo

sure

Cad

miu

mH

um

anch

ron

ic0.

005

Sig

nifi

can

tpr

otei

nu

ria

1010

11

11

15×

10−4

Hig

hN

.A.

expo

sure

sfr

oma

vari

ety

ofst

udi

esN

.A.7

N.A

.In

suffi

cien

tin

form

atio

nex

ists

N.A

.N

.A.

inth

eIR

ISfi

leto

mak

ean

yde

term

inat

ion

ofan

RfD

Ch

lorp

yrif

osH

um

anex

peri

men

tal

0.03

Pla

sma

chol

ines

tera

se10

101

11

11

10−3

Med

ium

30ca

psu

lein

hib

itio

nD

og2-

year

feed

ing

0.01

Dog

plas

ma

and

RB

C10

010

101

11

11×

10−4

Med

ium

chol

ines

tera

sein

hib

itio

n4,

6-D

init

ro-o

-H

um

anan

ecdo

tal

2.0

(L)8

Cat

arac

tfo

rmat

ion

1000

101

1010

11

10−3

Low

Eit

her

0.03

cycl

ohex

yl-

clin

ical

ther

apy

(cat

arac

t)or

0.07

phen

ol(g

row

thde

pres

sion

Rat

6-m

onth

feed

ing

64av

erag

edC

atar

act

form

atio

n9

1000

1010

110

11

10−2

Low

&sw

elli

ng)

Rat

6-m

onth

feed

ing

32av

erag

edG

row

thra

tede

pres

sion

,clo

udy

1000

1010

110

11

10−2

Low

swel

lin

gof

live

rce

lls

USING HUMAN DATA TO PROTECT THE PUBLIC’S HEALTH 239

,4

-Din

itro

phen

olH

um

anan

ecdo

tal

2.0

(L)

Cat

arac

tfo

rmat

ion

1000

101

1010

11

10−3

Low

Eit

her

0.02

clin

ical

ther

apy

(cat

arac

t)or

0.4

Rat

6-m

onth

feed

ing

130

Cat

arac

tfo

rmat

ion

1000

1010

110

11

10−1

Low

(gro

wth

depr

essi

onav

erag

ed&

swel

lin

g)

Rat

6-m

onth

feed

ing

5G

row

thra

tede

pres

sion

,clo

udy

1000

1010

110

11

10−3

10L

owsw

elli

ng

ofli

ver

cell

sth

eph

onH

um

anex

peri

men

tal

0.5

(L)

Pla

sma

chol

ines

tera

se10

010

110

11

15×

10−3

Low

6or

alex

posu

rein

hib

itio

nD

og2-

year

feed

ing

0.75

(L)

Pla

sma

chol

ines

tera

se10

0010

1010

11

18×

10−4

Low

inh

ibit

ion

thio

nH

um

anex

peri

men

tal

0.05

11P

lasm

ach

olin

este

rase

100

101

101

11

10−4

Med

ium

8sh

ort

term

inh

ibit

ion

Dog

90-d

ayor

alex

posu

re0.

06(L

)P

lasm

ach

olin

este

rase

1000

1010

101

11

10−5

Med

ium

inh

ibit

ion

luor

ine

Hu

man

epid

emio

logy

0.06

Obj

ecti

onab

lede

nta

lflu

oros

is1

11

11

11

10−2

Hig

hN

.A.

(sol

ubl

eN

.A.

N.A

.In

suffi

cien

tin

form

atio

nex

ists

N.A

.N

.A.

flu

orid

e)in

the

IRIS

file

tom

ake

any

dete

rmin

atio

nof

RfD

alat

hio

nH

um

anex

peri

men

tal

0.23

Ery

thro

cyte

chol

ines

tera

se10

101

11

11

10−2

Med

ium

0.4

feed

ing

inh

ibit

ion

Rat

2-ye

arfe

edin

g5

Dec

reas

edbr

ain

chol

ines

tera

se10

010

101

11

15×

10−2

Med

ium

and

decr

ease

dbo

dyw

eigh

tan

gan

ese

Hu

man

data

ofse

vera

l0.

14N

oL

OA

EL

give

n,C

NS

effe

cts

11

11

11

11.

10−3

Med

ium

N.A

.ty

pes

appe

arto

occu

rat

hig

her

dose

sN

.A.

N.A

.IR

ISst

ates

that

hu

man

data

are

N.A

.N

.A.

supe

rior

toan

yda

taob

tain

edfr

oman

imal

toxi

city

stu

dies

asth

eba

sis

ofan

RfD

,sin

ceth

eph

ysio

logi

cre

quir

emen

tsfo

rm

anga

nes

eva

ryam

ong

spec

ies

eth

ylm

ercu

ryH

um

anep

idem

iolo

gica

l0.

0011

(B)

Infa

nt

deve

lopm

enta

l10

31

11

31

10−4

Med

ium

5po

ison

ing

neu

rolo

gica

labn

orm

alit

ies

Rat

deve

lopm

enta

l0.

005

Dec

reas

ein

oper

ant

beh

avio

r30

010

101

13

12×

10−5

Med

ium

gava

gean

d26

-mon

thpe

rfor

man

ce,r

edu

ced

body

feed

ing

stu

dyw

eigh

tga

ins,

min

imal

clin

ical

sign

sof

neu

roto

xici

ty,k

idn

eyda

mag

eol

ybde

nu

mH

um

an0.

14(L

)In

crea

sed

uri

cac

id30

31

101

11

10−3

Med

ium

2ep

idem

iolo

gica

ldi

etar

yR

at13

-wee

kfe

edin

g2.

5(L

)R

etar

ded

wei

ght

gain

1000

1010

33

11

10−3

Low

itra

teH

um

anep

idem

iolo

gy1.

6E

arly

clin

ical

sign

sof

11

11

11

11.

6H

igh

8su

rvey

sm

eth

emog

lobi

nem

ia<

10%

Rat

24-m

onth

drin

kin

g20

12D

ilat

edbr

onch

i,fi

bros

is,a

nd

100

1010

11

11

10−1

Med

ium

wat

erem

phys

ema

2 E E F M M M M N

240

DOURSON ET AL.

TA

BL

E1–

Con

tin

ued

Ch

emic

aln

ame

NO

AE

L,

Un

cert

ain

tyfa

ctor

3R

fDra

tio:

(as

onE

PA

’sL

OA

EL

orR

fDH

um

anto

IRIS

)S

peci

e/T

ype

ofst

udy

BM

D2

Cri

tica

leff

ect(

s)T

otal

HA

LS

DM

FR

fDco

nfi

den

cean

imal

Nit

rite

Hu

man

epid

emio

logy

1.013

Ear

lycl

inic

alsi

gns

of1

11

11

110

10−1

Hig

h5

surv

eys

met

hem

oglo

bin

emia

l<10

%R

at24

-mon

thdr

inki

ng

2D

ilat

edbr

onch

i,fi

bros

is,a

nd

100

1010

11

11

10−2

Med

ium

wat

erem

phys

ema

Pir

imip

hos

-H

um

an56

-day

0.25

Tra

nsi

ent

plas

ma

2510

12.

51

11

10−2

Hig

hE

ith

er2

met

hyl

expe

rim

enta

lfee

din

gch

olin

este

rase

inh

ibit

ion

(rat

)or

20R

at2-

year

feed

ing

0.5

Pla

sma

chol

ines

tera

se10

010

101

11

15×

10−3

Hig

h(d

og)

inh

ibit

ion

Dog

2-ye

arfe

edin

g0.

5(L

)B

rain

chol

ines

tera

sein

hib

itio

n10

0010

1010

11

15×

10−4

Med

ium

Sel

eniu

man

dH

um

anfo

odan

dso

il0.

015

Cli

nic

alse

len

osis

33

11

11

15×

10−3

Hig

h6

com

pou

nds

epid

emio

logy

Rat

deve

lopm

enta

l0.

075

50%

redu

ctio

nin

nu

mbe

rof

100

1010

11

11

10−4

Hig

hto

xici

tyyo

un

gre

ared

Sil

ver

Hu

man

anec

dota

l0.

014

Arg

yria

33

11

11

15×

10−3

Low

0.2

stu

dies

Rat

218-

day

drin

kin

g89

(L)

Ven

tric

ula

rh

yper

trop

hy

3000

1010

103

11

10−2

Low

wat

erex

posu

re1,

1,2-

Tri

chlo

ro-

Hu

man

occu

pati

onal

273

Psy

chom

otor

impa

imen

t10

101

11

11

101

Low

N.A

.1,

2,2-

trifl

uor

o-ex

posu

reet

han

eN

.A.

N.A

.In

suffi

cien

tin

form

atio

nex

ists

N.A

.N

.A.

inth

eIR

ISfi

leto

mak

ean

yde

term

inat

ion

ofR

fDW

arfa

rin

Hu

man

expe

rim

enta

l0.

029

Incr

ease

dpr

oth

rom

bin

tim

e10

010

110

11

13×

10−4

Low

N.A

.In

appr

opri

ate

N.A

.IR

ISst

ates

‘Bec

ause

ofN

.A.

N.A

.m

arke

ddi

ffer

ence

sin

the

susc

epti

bili

tyof

diff

eren

tsp

ecie

sto

the

effe

cts

ofw

arfa

rin

,it

wou

ldbe

inap

prop

riat

eto

deri

vean

RfD

from

stu

dies

onlo

wer

anim

als”

E PUBLIC’S HEALTH 241

USING HUMAN DATA TO PROTECT THZ

inc

and

Hu

man

expe

rim

enta

l59

.7(L

)D

ecre

ase

iner

yth

rocy

te3

11

31

11

10−1

Med

ium

N.A

.co

mpo

un

dsdi

etsu

pple

men

tsu

pero

xide

dism

uta

seco

nce

ntr

atio

nin

adu

lts

N.A

.N

.A.

Insu

ffici

ent

info

rmat

ion

exis

tsN

.A.

N.A

.in

the

IRIS

file

tom

ake

any

dete

rmin

atio

nof

RfD

1A

llda

tafr

omw

hic

hw

ees

tim

ate

anim

al-b

ased

RfD

sar

eta

ken

from

EP

A’s

IRIS

un

less

oth

erw

ise

stat

ed.J

udg

men

tsof

con

fide

nce

leve

lsfo

ran

imal

-bas

edri

skva

lues

wer

eba

sed

onM

.Dou

rson

’sex

peri

ence

wit

hth

eR

fD/R

fCW

ork

Gro

up

ofE

PA

.2

All

valu

esar

ein

mg/

kg-d

ayan

dar

eN

OA

EL

su

nle

ssot

her

wis

est

ated

:(L

),L

OA

EL

;(B

),be

nch

mar

kdo

se(B

MD

).3

Un

cert

ain

tyfa

ctor

sar

eH

,ave

rage

hu

man

tose

nsi

tive

hu

man

;A,a

nim

alto

hu

man

;L,L

OA

EL

toN

OA

EL

:S,s

ubc

hro

nic

expo

sure

toch

ron

ic;D

,dat

abas

ein

suffi

cien

cy;M

F,m

odif

yin

gfa

ctor

toac

cou

nt

for

un

cert

ain

itie

sn

otco

vere

dby

the

trad

itio

nal

fact

ors.

Not

eth

atfo

rth

isex

erci

se,t

he

Dan

dM

Ffa

ctor

sw

ere

con

side

red

tobe

the

sam

ebe

twee

nth

ean

imal

-bas

edR

fDan

dth

eh

um

an-b

ased

RfD

,sin

ceth

eyor

igin

ated

from

the

sam

eda

taba

sew

ith

one

exce

ptio

n(n

itri

te)

sin

cein

form

atio

non

IRIS

allo

wed

adi

ffer

ence

.Th

ese

fact

ors

may

orm

ayn

otbe

the

sam

eu

pon

reev

alu

atio

nof

new

erto

xici

tyda

tan

oton

IRIS

.4

Bot

hth

edo

gan

dh

um

anst

udi

essh

owed

plas

ma

chol

ines

tera

sein

hib

itio

nat

the

low

est

dose

.F

orth

isco

mpa

riso

n,

both

low

dose

sw

ere

con

side

ras

NO

AE

Ls;

ifth

eyar

eco

nsi

dere

das

LO

AE

Ls,

the

RfD

sm

igh

tbe

low

erfo

rbo

th,b

ut

the

rati

obe

twee

nth

ese

RfD

sw

ould

beth

esa

me.

5D

ata

deri

ved

from

Oko

jiet

al.(

1999

).6

IRIS

stat

es“t

hat

labo

rato

ryan

imal

sar

ein

appr

opri

ate

mod

els

for

stu

dyin

gth

eto

xici

tyof

ben

zoic

acid

inh

um

ans...

,”bu

tth

engo

onto

give

anR

fDof

10−2

wit

ha

furt

her

com

men

tth

atit

“is

not

appr

opri

ate

tou

seth

ean

imal

data

for

the

RfD

.”7

N.A

.,n

otap

plic

able

.8

Bas

edon

anal

ogy

to2,

4-di

nit

roph

enol

.9

IRIS

stat

esth

at“t

he

subc

hro

nic

diet

ary

rat

stu

dy...

isn

otu

sefu

lfor

the

RfD

deri

vati

onbe

cau

seex

peri

men

tala

nim

als

are

not

adeq

uat

em

odel

sfo

rth

ein

duct

ion

ofca

tara

cts

inh

um

ans

bylo

w-l

evel

oral

expo

sure

todi

nit

roph

enol

s,an

d,th

eref

ore,

anR

fDba

sed

onth

era

tda

tam

ayn

otbe

adeq

uat

ely

prot

ecti

vefo

rh

um

ans.

”T

his

com

men

tis

not

fou

nd

onth

e2,

4-di

nit

roph

enol

IRIS

file

,bu

tap

plie

sto

itas

wel

l.10

Val

ue

give

non

IRIS

.Ple

ase

not

eth

eco

mm

ent

abov

eon

4,6-

din

itro

-o-c

yclo

hex

ylph

enol

.11

EP

A’s

IRIS

list

sth

isva

lue

asa

NO

EL

,bu

tal

soad

dsan

un

cert

ain

tyfa

ctor

for

LO

AE

Lto

NO

AE

Lex

trap

olat

ion

beca

use

ofth

epr

oxim

ity

ofth

isN

OE

Lto

brai

nch

olin

este

rase

inh

ibit

ion

indo

gsat

0.71

mg/

kg-d

ay.

12B

ased

onth

eto

xici

tyof

nit

rite

wit

ha

10-f

old

incr

ease

inth

eN

OA

EL

.13

Bas

edon

the

toxi

city

ofn

itra

tew

ith

anad

just

men

tto

this

dose

wit

ha

10-f

old

MF.

242 DOURSON ET AL.

responding animal-based RfD or RfC.

3 These frequency values are based on ratios that fall within a 10-fold range of each other, for example 0.3 mg/kg-day≤RfD≤ 0.3 mg/kg-day. The use of such a range is consistent with the definition of RfDsand RfCs in that “uncertainty spans perhaps an order of magnitude”and thus their expected level of precision. However, the precision ofrisk values has never been explicitly addressed in EPA risk values (seeFelter and Dourson, 1998, for more discussion of this), and thereforethe range we use here is only for demonstration. Other equally valid

concentrations, net exposures as area under the concen-tration curves, that is believed to be most closely asso-ciated with the toxic effects. Any narrative describingthe mode of action of a compound should also convey theform of the chemical believed to be responsible for initi-ating the cascade of steps leading to the toxic responses.Similarly, extrapolation between species should be con-ducted using a dose metric that reflects this under-standing of the mode of action of the compound.

For example, a mode of action statement for vinylchloride would be: tumors associated with vinyl chlo-ride are caused by mutational effects arising from thereaction of epoxide metabolites with DNA resulting inincreases in the mutational frequency in specific cells.The dose metric for this mode of action would be a mea-sure of the net tissue exposure of the epoxide metabo-lites (EPA, 2000b). For chloroform, the mode of actionstatement would read: tumors in the liver and kidneyinduced by chloroform arise due to mutations duringrecurrent episodes of cytotoxicity and cell proliferationassociated with metabolism of chloroform to phosgenein cells with high CYP 2E1 activity. Here the dose metricwould be related to peak rates of metabolism of chloro-form in liver and in kidney cortex (ILSI, 1997).

Pharmacokinetic and dosimetry data in risk assess-ment. Several advances in dose–response assessmenthave occurred over the past 20 years due to increasedemphasis on understanding modes of action and on de-termining the major biological determinants that con-tribute to pharmacokinetic behavior in animals andhumans. Before 1980, it was common to simply col-lect pharmacokinetic data and analyze it empiricallywith mathematical models to evaluate the correspon-dence of the data with specific compartmental models.Sometimes the data were not analyzed at all. A diffi-culty with these compartmental approaches was in ex-trapolation of the results to important untested situ-ations. Even when impressive fits to animal data wereobtained, how confident could we be that the compoundswould have similar kinetic behavior in humans? If hu-man data were actually obtained from volunteers, howconfident could we be that the data obtained from asmall group of healthy individuals were representativeof pharmacokinetic behaviors expected in a larger hu-man population? These two questions led to increasedemphasis on development of physiologically based phar-macokinetic (PBPK) models in which the organism isdescribed realistically in terms of anatomy, physiol-ogy, biochemical parameters of distribution, and thephysical-chemical characteristics of the test compound(Gerlowski and Jain, 1983). These particular models aremore amenable to extrapolation from test animals tohumans and are also amenable to establishing the im-portance of variability within the human population by

Monte Carlo methods.

RESULTS

Comparison of Human- and ExperimentalAnimal-Based RfDs and RfCs

Table 1 shows a comparison of all RfDs based on hu-man data as found on IRIS (EPA, 2000b) or for sev-eral chemicals on ITER (TERA, 2000), and those weestimate from experimental animal data found on IRISusing the criteria defined above. For 36% (8 of 22 com-parisons), the human- and animal-based RfDs are com-parable, that is within the limits of their correspond-ing precision.3 For 23% (5 of 22 comparisons), the RfDsbased on human data are lower than the correspondingRfDs based on animal data. For 41% (9 of 22 compar-isons), RfDs based on animal data are lower than thosebased on human data. An animal-based RfD could notbe estimated in 6 of 28 possible comparisons, since ani-mal information was judged to be either insufficient orirrelevant.

Table 2 shows a comparison of all RfCs based on hu-man data as found on IRIS (EPA, 2000b) or for sev-eral chemicals on ITER (TERA, 2000), and those weestimate from experimental animal data found on IRISusing criteria defined above. For 45% (5 of 11 compar-isons), the RfCs are comparable, that is within the lim-its of their corresponding precision.3 In no cases werethe RfCs based on human data lower than the corre-sponding RfCs based on animal data within the limitsof precision being assumed. For 55% (6 of 11 compar-isons), RfCs based on animal data are lower than thosebased on human data. An animal-based RfC could notbe estimated for 4 of 15 times, since animal informationwas judged to be either insufficient or irrelevant.

Figure 1 shows a frequency plot of human- to exper-imental animal-based RfD or RfC ratios from Tables 1and 2.

Differences in the ratios of the human to the exper-imental animal RfD or RfC can also be shown as thenumber that were above or below a value of 1 withoutregards to any considerations of precision. Here val-ues below 1 indicate that human data resulted in alower RfD or RfC than animal data. For RfDs this fre-quency is 9 of 22 (or 41%). For RfCs this frequency is 3of 11 (or 27%). Collectively, this frequency is 12 of 33(or 36%). This percentage represents the number oftimes a human-based RfD or RfC was lower than a cor-

ranges may be determined.

USING HUMAN DATA TO PROTECT THE PUBLIC’S HEALTH 243

FIG. 1. Frequency of human- to animal-based RfD and RfC ratio from Tables 1 and 2. Ranges defined by logarithmic midpoints (e.g.,0.3 < 1 ≤ 3). Value of 1 indicates that animal-based and human-based risk values are the same.

Table 3 shows human to animal RfD ratios fromTable 1, where experimental or anecdotal clinicalhuman data formed the basis of EPA’s assessment. Thepurpose of this subset analysis is to determine whetherdifferent kinds of human data allow the development ofdifferent conclusions. Eight of 15 comparisons (53%) ofhuman to animal RfDs are less than 1. If anecdotal clin-ical data are excluded from these comparisons, then 3 of10 comparisons (30%) of experimental human-based toanimal-based RfDs are less than 1. Unfortunately toofew comparisons exist in order to make any definitiveconclusions on the relative proportions of these ratiosamongst the different types of human data as describedin the introduction. However, these percentages of ra-tios less than 1 found in Table 3 are not dissimilar fromthe percentage of ratios with the overall RfD databaseof 41% (found in Tables 1 and 2).

It is of interest to study the reasons why certain riskvalues could not be based on animal data. This occurredwith a frequency of 6 of 28 (or 21%) for RfDs and 4 of15 (or 27%) for RfCs. Table 4 shows the stated reasonsderived from Tables 1 and 2. For 40%, the stated reasonis a scientific judgment that the animal data are not rel-evant to the development of a risk value for humans asfurther explained in individual chemical files on IRIS(EPA, 2000b). For 60%, the animal data was not suffi-ciently described on EPA’s IRIS to develop a risk value.

Comparison of Human and ExperimentalAnimal Pharmacokinetics

Human data for risk assessment purposes includein vitro determinations of biochemical constants for

metabolism and tissue partition coefficients, limited

pharmacokinetic studies in specific human populations,and mechanistic studies using human tissues or humancells in vitro. The emphasis in this section is on the useof kinetic data and in vitro methods for assessing bi-ological determinants of kinetics in human tissues. AContemporary Concepts in Toxicology Workshop underthe auspices of the Society of Toxicology (SOT) TaskForce to Improve the Scientific Basis of Risk Assess-ment was held in September 1999. The report of theworkshop outlining the uses of human tissues in riskassessment is in press (MacGregor et al., 2000). Thispaper provides a larger perspective on use of humantissues for mechanistic studies.

We describe a series of examples to illustrate the useof human pharmacokinetic data in risk assessment.

Dichloromethane

Human equivalent concentrations. Occupationaltoxicology has historically featured the use of humanvolunteers for assessing the pharmacokinetics ofindustrial chemicals. Long before human testing wasundertaken, the occupational importance and history ofthe compound’s usage in industrial environments wasestablished. In general, exposure standards had beenestablished and the human exposures were conductedat exposure levels at or below existing ThresholdLimit Values or equivalent occupational standards.Work with human volunteers in the 1970’s establishedthe conversion of dichloromethane (DCM; methylenechloride) to carbon monoxide and the production ofrelatively high levels of carboxyhemoglobin (HbCO)—from 5 to 20%—following high level exposures to this

solvent. The analytical methods for measuring most

244 DOURSON ET AL.

TA

BL

E2

Su

mm

ary

ofU

.S.E

PA

’sR

fCs

onIR

ISas

ofM

ay20

001

Hu

man

Equ

ival

ent

Ch

emic

aln

ame

NO

AE

L,

Un

cert

ain

tyfa

ctor

3R

fCra

tio:

(as

onE

PA

’sS

peci

e/T

ype

LO

AE

L,o

rR

fCH

um

anto

IRIS

)of

stu

dyB

MD

2C

riti

cale

ffec

t(s)

Tot

alH

AL

SD

MF

RfC

con

fide

nce

anim

al

Am

mon

iaH

um

anoc

cupa

tion

al2.

3L

ack

ofev

iden

ceof

decr

ease

d30

101

11

31

10−1

Med

ium

20pu

lmon

ary

fun

ctio

nor

chan

ges

insu

bjec

tive

sym

ptom

atol

ogy

Rat

subc

hro

nic

1.9

(L)

Incr

ease

dse

veri

tyof

rhin

itis

and

300

103

314

31

10−3

Med

ium

inh

alat

ion

pneu

mon

iaw

ith

resp

irat

ory

lesi

ons

Ber

ylli

um

Hu

man

occu

pati

onal

0.00

02(L

)B

eryl

liu

mse

nsi

tiza

tion

and

101

13

31

12×

10−5

Med

ium

N.A

.an

dco

mm

un

ity

prog

ress

ion

toch

ron

icex

posu

rebe

ryll

ium

dise

ase

(CB

D)

N.A

.5N

.A.

No

labo

rato

ryan

imal

mod

elN

.A.

N.A

.fu

lly

mim

ics

allf

eatu

res

ofh

um

anC

BD

Bro

mom

eth

ane

Hu

man

occu

pati

onal

2.2

(L)6

Neu

rolo

gica

leff

ects

100

101

101

11

10−2

N.A

.4

Rat

29-m

onth

0.48

(L)

Deg

ener

ativ

ean

dpr

olif

erat

ive

100

103

31

11

10−3

Hig

hin

hal

atio

nle

sion

sof

the

olfa

ctor

yep

ith

eliu

mof

the

nas

alca

vity

Car

bon

disu

lfide

Hu

man

occu

pati

onal

19.7

(B)

Per

iph

eral

ner

vou

ssy

stem

303

11

33

17×

10−1

Med

ium

8dy

sfu

nct

ion

Rat

90-d

ay27

Per

iph

eral

ner

vou

ssy

stem

300

33

110

31

10−2

Med

ium

inh

alat

ion

dysf

un

ctio

nC

hro

miu

m(a

cid

Hu

man

subc

hro

nic

0.00

0714

(L)

Nas

alse

ptu

mat

roph

y90

101

33

11

10−6

Low

N.A

.m

ists

and

aero

sols

)oc

cupa

tion

alN

.A.

N.A

.E

xper

imen

tala

nim

alst

udi

esN

.A.

N.A

.h

ave

not

repo

rted

onn

asal

mu

cosa

leff

ects

foll

owin

gin

hal

atio

nex

posu

res

N,N

-Dim

eth

ylfo

rmam

ide

Hu

man

occu

pati

onal

7.9

(L)

Dig

esti

vedi

stu

rban

cean

d30

010

13

33

13×

10−2

Med

ium

4m

inim

alh

epat

icch

ange

ssu

gges

tli

ver

effe

cts

Rat

4-w

eek

25(L

)H

epat

icce

llch

ange

s30

0010

310

73

31

10−3

Med

ium

inh

alat

ion

n-H

exan

eH

um

anin

hal

atio

n73

(L)

Neu

roto

xici

ty;

300

101

101

31

10−1

Med

ium

0.5

epid

emio

logy

elec

trop

hys

iolo

gica

lalt

erat

ion

sM

ouse

90-d

ay38

Epi

thel

iall

esio

ns

inth

e10

010

31

13

14×

10−1

Med

ium

inh

alat

ion

nas

alca

vity

245

USING HUMAN DATA TO PROTECT THE PUBLIC’S HEALTHyd

roge

ncy

anid

eH

um

anoc

cupa

tion

al2.

5(L

)C

NS

sym

ptom

san

dth

yroi

d10

0010

110

33

13×

10−3

Low

N.A

.ef

fect

sN

.A.

N.A

.In

suffi

cien

tin

form

atio

nex

ists

N.A

.N

.A.

inth

eIR

ISfi

leto

mak

ean

yde

term

inat

ion

ofR

fCan

gan

ese

Hu

man

occu

pati

on0.

05Im

pair

men

tof

neu

robe

hav

iora

l10

0010

110

33

15×

10−5

Med

ium

N.A

.ex

posu

refu

nct

ion

N.A

.N

.A.

IRIS

stat

esth

atan

imal

toxi

city

N.A

.N

.A.

data

qual

itat

ivel

ysu

ppor

tth

eh

um

anst

udi

esu

sed

asa

basi

sof

the

RfC

;qu

anti

fied

com

pari

son

sw

ere

not

prov

ided

ercu

ry,e

lem

enta

lH

um

anoc

cupa

tion

al0.

009

(L)

Han

dtr

emor

;in

crea

ses

inm

emor

y30

31

31

31

10−4

Med

ium

0.4

dist

urb

ance

s;sl

igh

tsu

bjec

tive

and

obje

ctiv

eev

iden

ceof

auto

nom

icdy

sfu

nct

ion

Rat

72-w

eek

inh

alat

ion

0.07

No

his

topa

thol

ogic

alev

iden

ce10

010

31

13

17×

10−4

Low

ofre

spir

ator

yda

mag

e;h

igh

erdo

ses

cau

sed

deve

lopm

enta

lto

xici

tyin

adi

ffer

ent

stu

dy

tyre

ne

Hu

man

occu

pati

onal

34C

NS

effe

cts

303

11

33

11

Med

ium

50M

ouse

subc

hro

nic

74(L

)B

ron

chio

lar

rege

ner

atio

n,

3000

103

103

31

10−2

Med

ium

inh

alat

ion

olfa

ctor

ym

etap

lasi

aan

dde

gen

erat

ion

,in

crea

sed

lun

gw

eigh

t,in

crea

sed

epit

hel

ial

hyp

erpl

asia

ofth

efo

rest

omac

het

rach

loro

eth

ylen

eH

um

anoc

cupa

tion

al24

(L)9

Incr

ease

dre

acti

onti

mes

to10

010

110

11

N.A

.2×

10−1

8N

.A.

0.6

sim

ple

neu

rolo

gica

ltes

tsM

ouse

103-

wee

kin

hal

atio

n36

3(L

)11R

edu

ced

surv

ival

(in

mal

es),

1000

1010

101

1N

.A.

3.6×

10−1

9N

.A.

hep

atot

oxic

ity

(mal

es),

lun

gco

nge

stio

nan

dn

eph

roto

xici

ty(m

ales

and

fem

ales

)ol

uen

eH

um

anoc

cupa

tion

al11

9(L

)N

euro

logi

cale

ffec

ts30

010

110

13

14×

10−1

Med

ium

5R

at2-

year

79(L

)D

egen

erat

ion

ofn

asal

1000

103

101

31

10−2

Med

ium

inh

alat

ion

epit

hel

ium

olu

ene

diis

ocya

nat

eH

um

anoc

cupa

tion

al0.

002

Ch

ron

iclu

ng

fun

ctio

nde

clin

e30

101

13

11

10−5

Med

ium

2m

ixtu

reT

DI)

Rat

chro

nic

0.01

(L)

Nec

roti

crh

init

isw

ith

epit

hel

ial

300

103

101

11

10−5

Med

ium

inh

alat

ion

atro

phy,

met

apla

sia,

and

infl

amm

atio

n

H M M S T T T (

246 DOURSON ET AL.

TA

BL

E2–

Con

tin

ued

Hu

man

Equ

ival

ent

Ch

emic

aln

ame

NO

AE

L,

Un

cert

ain

tyfa

ctor

3R

fCra

tio:

(as

onE

PA

’sL

OA

EL

,or

RfC

Hu

man

toIR

IS)

Spe

cie/

Typ

eof

stu

dyB

MD

2C

riti

cale

ffec

t(s)

Tot

alH

AL

SD

MF

RfC

con

fide

nce

anim

al

Xyl

enes

Hu

man

occu

pati

onal

60.7

6(L

)9S

ubj

ecti

vesy

mpt

oms

100

101

101

1N

.A.

10−1

10N

.A.

3R

atin

hal

atio

n17

7(L

)11F

etal

toxi

city

1000

1010

101

1N

.A.

1.8×

10−1

N.A

.de

velo

pmen

tal

toxi

city

1A

llda

tafr

omw

hic

hw

ees

tim

ate

anim

al-b

ased

RfC

sar

eta

ken

from

EP

A’s

IRIS

un

less

oth

erw

ise

stat

ed.J

udg

men

tsof

con

fide

nce

leve

lsfo

ran

imal

-bas

edri

skva

lues

wer

eba

sed

onM

.Dou

rson

’sex

peri

ence

wit

hth

eR

fD/R

fCW

ork

Gro

up

ofE

PA

.2

All

valu

esar

eh

um

aneq

uiv

alen

tco

nce

ntr

atio

nin

mg/

m3

and

are

NO

AE

Ls

un

less

oth

erw

ise

stat

ed:(

L),

LO

AE

L;(

B),

ben

chm

ark

con

cen

trat

ion

(BM

C).

3U

nce

rtai

nty

fact

ors

are

H,a

vera

geh

um

anto

sen

siti

veh

um

an;A

,an

imal

toh

um

an;L

,LO

AE

Lto

NO

AE

L;S

,su

bch

ron

icex

posu

reto

chro

nic

;D,d

atab

ase

insu

ffici

ency

;MF,

mod

ifyi

ng

fact

orto

acco

un

tfo

ru

nce

rtai

nti

esn

otco

vere

dby

the

trad

itio

nal

fact

ors.

Not

eth

atfo

rth

isex

erci

se,t

he

Dan

dM

Ffa

ctor

sw

ere

con

side

red

tobe

the

sam

ebe

twee

nth

ean

imal

-bas

edR

fDan

dth

eh

um

an-b

ased

RfD

,sin

ceth

eyor

igin

ated

from

the

sam

eda

taba

se.T

hes

efa

ctor

sm

ayn

otbe

the

sam

eu

pon

reev

alu

atio

nof

new

erto

xici

tyda

tan

oton

IRIS

.4

Th

eva

lue

ofth

issu

bch

ron

icto

chro

nic

un

cert

ain

tyfa

ctor

is1;

itis

subs

um

edin

the

desc

ript

ion

ofth

eu

nce

rtai

nty

fact

orfo

rda

taba

sede

fici

enci

esas

per

IRIS

.5N

.A.,

not

appl

icab

le.

6V

alu

eis

from

AT

SD

Ran

dth

ere

sult

ing

risk

valu

eis

am

inim

alri

skle

vel(

MR

L).

Som

edi

ffer

ence

sex

ist

amon

gm

eth

ods

toes

tim

ate

thes

eri

skva

lues

amon

gE

PA

and

AT

SD

R.

Ple

ase

see

ww

w.t

era.

org/

iter

/met

hod

s.S

peci

fic

valu

esof

AT

SD

Ran

dE

PA

can

befo

un

dat

ww

w.t

era.

org/

iter

un

der

“bro

mom

eth

ane.

”7

An

eval

uat

ion

ofth

eor

igin

alst

udy

wit

hin

the

over

alld

atab

ase

may

allo

wa

redu

ctio

nin

this

fact

orto

ava

lue

of3.

8S

peci

fic

valu

esof

AT

SD

Ran

dH

ealt

hC

anad

aca

nbe

fou

nd

atw

ww

.ter

a.or

g/it

eru

nde

r“t

etra

chlo

roet

hyl

ene.

”9

Val

ue

isfr

omA

TS

DR

and

the

resu

ltin

gri

skva

lue

isa

min

imal

risk

leve

l(M

RL

).S

ome

diff

eren

ces

exis

tam

ong

met

hod

sto

esti

mat

eth

ese

risk

valu

esam

ong

EP

A,A

TS

DR

,an

dH

ealt

hC

anad

a.P

leas

ese

ew

ww

.ter

a.or

g/it

er/m

eth

ods.

10S

peci

fic

valu

esof

AT

SD

Ran

dH

ealt

hC

anad

aca

nbe

fou

nd

atw

ww

.ter

a.or

g/it

eru

nde

r“x

ylen

es.”

11V

alu

eis

from

Hea

lth

Can

ada

and

the

resu

ltin

gri

skva

lue

isa

tole

rabl

eco

nce

ntr

atio

n(T

C).

Som

edi

ffer

ence

sex

ist

amon

gm

eth

ods

toes

tim

ate

thes

eri

skva

lues

amon

gE

PA

and

AT

SD

R.P

leas

ese

ew

ww

.ter

a.or

g/it

er/m

eth

ods.

USING HUMAN DATA TO PROTECT THE PUBLIC’S HEALTH 247

TA

BL

E3

Su

mm

ary

RfD

sfr

omT

able

1B

ased

onE

xper

imen

tal

orC

lin

ical

Hu

man

Dat

a

Ch

emic

aln

ame

rati

o:H

um

an(a

son

EP

A’s

IRIS

)S

peci

e/T

ype

ofst

udy

Cri

tica

leff

ect(

s)R

fDto

anim

al

Ald

icar

bH

um

anex

peri

men

talg

avag

eC

lin

ical

sign

sof

chol

ines

tera

sein

hib

itio

n,

10−3

Eit

her

1(c

lin

ical

sign

s)or

plas

ma

chol

ines

tera

sein

hib

itio

n3

(pla

sma

Ch

E)

Rat

deve

lopm

enta

lgav

age

Cli

nic

alsi

gns

ofch

olin

este

rase

inh

ibit

ion

10−3

Dog

52-w

eek

feed

ing

Pla

sma

chol

ines

tera

sein

hib

itio

n3×

10−4

Bar

ium

Hu

man

expe

rim

enta

l,In

crea

sed

bloo

dpr

essu

re7×

10−2

0.4

epid

emio

logi

cald

rin

kin

gw

ater

Rat

chro

nic

drin

kin

gw

ater

Incr

ease

dki

dney

wei

ght

10−1

Bay

gon

Hu

man

expe

rim

enta

lsin

gle

dose

Mil

dch

olin

ergi

csy

mpt

oms,

RB

C4×

10−3

0.8

chol

ines

tera

sein

hib

itio

nD

og12

-mon

thfe

edin

gC

hol

ines

tera

sein

hib

itio

n5×

10−3

Ch

lorp

yrif

osH

um

anex

peri

men

talc

apsu

leP

lasm

ach

olin

este

rase

inh

ibit

ion

10−3

30D

og2-

year

feed

ing

Pla

sma

and

RB

Cch

olin

este

rase

inh

ibit

ion

10−4

4,6-

Din

itro

-o-c

yclo

hex

ylph

enol

Hu

man

anec

dota

lcli

nic

alth

erap

yC

atar

act

form

atio

n2×

10−3

Eit

her

0.03

(cat

arac

t)R

at6-

mon

thfe

edin

gC

atar

act

form

atio

n6×

10−2

or0.

07(g

row

th)

depr

essi

onR

at6-

mon

thfe

edin

gG

row

thra

tede

pres

sion

,clo

udy

swel

lin

gof

10−2

and

swel

lin

g)li

ver

cell

s2,

4-D

init

roph

enol

Hu

man

anec

dota

lcli

nic

alth

erap

yC

atar

act

form

atio

n2×

10−3

Eit

her

0.02

(cat

arac

t)or

0.4

Rat

6-m

onth

feed

ing

Cat

arac

tfo

rmat

ion

10−1

(gro

wth

depr

essi

onan

dR

at6-

mon

thfe

edin

gG

row

thra

tede

pres

sion

,clo

udy

swel

lin

gof

10−3

swel

lin

g)li

ver

cell

sE

thep

hon

Hu

man

expe

rim

enta

lora

lexp

osu

reP

lasm

ach

olin

este

rase

inh

ibit

ion

10−3

6D

og2-

year

feed

ing

Pla

sma

chol

ines

tera

sein

hib

itio

n8×

10−4

Eth

ion

Hu

man

expe

rim

enta

lsh

ort

term

Pla

sma

chol

ines

tera

sein

hib

itio

n5×

10−4

8D

og90

-day

oral

expo

sure

Pla

sma

chol

ines

tera

sein

hib

itio

n6×

10−5

Mal

ath

ion

Hu

man

expe

rim

enta

lfee

din

gE

ryth

rocy

tech

olin

este

rase

depr

essi

on2×

10−2

0.4

Rat

2-ye

arfe

edin

gD

ecre

ased

brai

nch

olin

este

rase

and

decr

ease

d5×

10−2

body

wei

ght

Pir

imip

hos

-met

hyl

Hu

man

56-d

ayex

peri

men

talf

eedi

ng

Tra

nsi

ent

plas

ma

chol

ines

tera

sein

hib

itio

n1×

10−2

Eit

her

2(r

at)

orR

at2-

year

feed

ing

Pla

sma

chol

ines

tera

sein

hib

itio

n5×

10−3

20(d

og)

Dog

2-ye

arfe

edin

gB

rain

chol

ines

tera

sein

hib

itio

n5×

10−4

Sil

ver

Hu

man

anec

dota

lstu

dies

Arg

yria

10−3

0.2

Rat

218-

day

drin

kin

gw

ater

Ven

tric

ula

rh

yper

trop

hy

10−2

expo

sure

War

fari

nH

um

anex

peri

men

tal

Incr

ease

dpr

oth

rom

bin

tim

e3×

10−4

Inap

prop

riat

eIR

ISst

ates

“Bec

ause

ofm

arke

ddi

ffer

ence

sin

N.A

.N

.A.

the

susc

epti

bili

tyof

diff

eren

tsp

ecie

sto

the

effe

cts

ofw

arfa

rin

,it

wou

ldbe

inap

prop

riat

eto

deri

vean

RfD

from

stu

dies

onlo

wer

anim

als”

Zin

can

dco

mpo

un

dsH

um

anex

peri

men

tald

iet

Dec

reas

ein

eryt

hro

cyte

supe

roxi

dedi

smu

tase

10−1

supp

lem

ent

N.A

.co

nce

ntr

atio

nin

adu

lts

N.A

.In

suffi

cien

tin

form

atio

nex

ists

inth

eIR

ISfi

leN

.A.

tom

ake

any

dete

rmin

atio

nof

RfD

248 DOURSON ET AL.

compounds in blood and excreta were new and manyhad only moderate sensitivity. The work with DCM es-tablished the exposure concentrations associated withspecific increases in HbCO and led to strategies for con-trolling DCM that insured no more than a 5% increasein blood HbCO. This early work with DCM, while quan-titative, provided simple mathematical relationships tocorrelate specific exposures with increased HbCO; theydid not really allow prediction of these values fromknowledge of human metabolism, physiology, and ac-tivity levels.

In the 1980s, DCM was found to be carcinogenic inmice, shifting the end point from HbCO to the pos-sibility that DCM might cause cancer in exposed in-dividuals. A risk assessment for DCM was proposed,based on a PBPK model, that estimated the tissueexposures in lung and liver to metabolites of DCM(Andersen et al., 1987). A variety of data indicatedthat the carcinogenicity in mice was most likely as-sociated with metabolites formed by conjugation ofDCM with glutathione, catalyzed by glutathione S-transferase (GST). This metabolic clearance pathwaycompetes with a higher affinity, though lower ca-pacity oxidation of DCM by cytochrome P450. Theisozyme involved is now recognized to be CYP2E1.The dose metric used for analysis in the region of ob-servation was the integrated production of the glu-tathione conjugate per volume tissue per day. Thissame dose metric was estimated for humans by usinga PBPK model that included physiological parametersfor humans and kinetic constants for the glutathione-S-transferase enzyme in lung and liver for both themouse and the human. Using model substrates, ac-tivities for the human liver and lung enzymes wereestimated in studies using microsomes obtained fromaccident victim’s tissues. Later work established theactivities of these enzymes toward DCM itself (Reitzet al., 1989).

Controlled pharmacokinetic studies had been con-ducted in human volunteers using several different con-centrations of DCM (Andersen et al., 1991). These stud-ies were evaluated with a PBPK model to assess theconsistency of predictions based on scaling the rodentPBPK model to predict actual human data. The ear-lier risk assessment had simply used a model with hu-man parameters to make predictions of the dose met-rics. With moderate refinements to the portion of themodel describing carbon monoxide distribution after itsformation by DCM oxidation, the model predicted theblood and exhaled air time-course for DCM. For CO,the model adequately predicted blood HbCO and ex-haled CO. The ability to match concentrations of DCM,HbCO, and CO in humans provides confidence that thescale-up from rats to humans is feasible. These resultscollectively and individually provide evidence for thecorrection in the numerator of the RfC, for example,

that which is required for developing a human equiva-

lent concentration. The dose metric used is the amountof HbCO formed for an occupational concern for car-boxyhemoglobin or the amount metabolized in lung orliver by the glutathione-S- transferase reactions. Thislatter dose metric is estimated from an accurate predic-tion of the blood concentrations of DCM and the enzymeactivities in the target tissues. These PBPK modelsallow estimation of the human exposure concentrationsthat are required to provide a tissue dose similar to thatassociated with the point of departure concentration inthe test animal.

Human variability. The denominator of the RfC andRfD includes uncertainty factors for interspecies and in-traindividual differences. The RfC methodology (EPA,1994) indicates that the interspecies factor can be re-duced when compound specific kinetic data are used toderive the human equivalent concentration. A secondapplication of these kinetic models is to estimate theimpact of variability in physiology, metabolism, etc. inpharmacokinetic variability within the human popula-tion. For this application, ranges of model parameters,such as breathing and blood flow rates, body compo-sition, and metabolic parameters, are introduced intothe model specification. The PBPK model is run mul-tiple times during which the parameters are selectedfrom these distributions by a sampling algorithm. Theresultant output is a distribution for target tissue doseexpected for a diverse population at the specified ex-posure concentration. By assessing the variability inthis distribution of tissue doses, it becomes possibleto decide if the factor of 10 for intraindividual differ-ences is adequate or if it needs to be adjusted. Ex-amples of use of Monte Carlo sampling of parameterdistributions to assess tissue dose with DCM have fo-cussed on standard setting for the workplace (Thomaset al., 1998) and for environmental exposures (Clewell,1996).

The work with DCM provides a template for applica-tions of human data in risk assessment based on tox-icity results in animals. Mechanistic studies usuallyset the foundation for assessing the dose metric thatis important to estimate as a measure of tissue dose.The PBPK model indicates the important parametersthat determine the dose of active forms of the com-pound at target tissues. Studies with human tissues,coupled with knowledge of human physiology/anatomy,provide a method to estimate human tissue doses ofactive compounds and a method to derive the HEC.Focused, limited, human-volunteer studies, develop invivo data that allows refinement and validation of thepredictions of the human PBPK model. The completedPBPK description lends itself to a refined, quantita-tive analysis of variability in pharmacokinetic behav-ior in the human population. This paradigm for useof human studies would be applicable to RfC, RfD or

cancer assessments for various end points. In contrast

USING HUMAN DATA TO PROTECT THE PUBLIC’S HEALTH 249

to the state-of-the-art in analytical chemistry in the1970s, many of the pharmacokinetic studies in humanscan now be conducted at much lower, almost tracer (ortrace) levels of contaminants mitigating concerns aboutpossible adverse effects of compounds in the humanvolunteers.

Vinyl Chloride

Pharmacokinetic (PK) modeling plays an increas-ingly accepted role in many of the efforts in the newIRIS Pilot project activities to integrate the risk as-sessment process for specific compounds. Vinyl chlo-ride (VC) documentation has been completed within thelast year (EPA, 2000b). The critical end points with VCare liver toxicity and liver cancer—hemangiosarcoma.VC is an animal and human carcinogen with strongsite-concordance between species. Mechanistic workhas firmly established a central role for an epox-ide, metabolite in causing DNA adducts, mutations,and tumors. Pharmacokinetic and disposition stud-ies have been conducted in multiple species of lab-oratory animals and limited pharmacokinetic uptakestudies were conducted with human volunteers at lowconcentrations. The IRIS document with VC includesa PBPK model to assess the consistency in dosime-try between animals and humans, and provides meth-ods for extrapolating across dose routes and betweenspecies. The human data provides a test of modelpredictions and reassurance that the standards setbased on these model estimates of dose are likely to besound.

Inhaled Acids and Esters

A wide variety of organic acids and esters have re-cently been associated with degeneration and toxicity inthe nasal olfactory epithelium in rats and mice. It wasunclear whether these compounds would have similareffects in humans. Within the past two to three years,PBPK models have been completed and published forseveral of these compounds—vinyl acetate (Plowchalket al., 1997), acrylic acid (Frederick et al., 1998), andmethyl methacrylate (Andersen et al., 1999). Dosime-try model development with these compounds has in-cluded evaluation of uptake from the rodent airstream,parameterization of nasal models for airflow and tissueuptake, and measurement of metabolizing enzymes inepithelial tissues throughout the nose. These dosime-try models predict differences between humans and ro-dents and support estimation of HECs for these com-pounds. A workshop was held in Research TrianglePark, North Carolina, in February 1998. Participantsdiscussed the utility of these dosimetry models for nasaldosimetry and risk assessment and continuing dataneeds. A major continuing data need enumerated was

for human studies of scrubbing of compounds from the

nasal airstream using specific breathing patterns andsampling strategies (Andersen and Jarabek, 2000). Thevalidation of existing models by selective, carefully de-signed human studies was recognized as an importantresearch priority.

Boric Acid

The most sensitive end point with boric acid in labora-tory animals is developmental toxicity in the offspringof Sprague–Dawley rats (Dourson et al., 1998). Thisinorganic acid has fairly straightforward pharmacoki-netic behavior. It is not metabolized to any appreciableextent; it is primarily excreted into the urine via the kid-neys; and it does not accumulate anywhere in the body.The main issue in assessing the uncertainty factors forthis compound is estimation of the net tissue exposureto fetuses in rats and any differences in exposure forequivalent doses in humans. General arguments canbe offered regarding the expected kinetic behavior. Forinstance, the volume of distribution should follow bodyweight (or at least lean body weight) and renal clear-ance should follow body weight raised to the 2/3 power(see, for example, NRC, 1986). These are theoretical ar-guments for generic compounds. It would be best to esti-mate these differences by direct determination of boricacid clearance studies in pregnant and nonpregnantfemales.

These renal clearance studies have recently been con-ducted by the University of California, Irvine (Vaziriet al., 2000). Murray and Andersen (2000) provide apreliminary outline of the results of these studies, in-cluding relative areas under the blood curve and thevariability observed in renal boric acid clearance in bothrats and women. These studies provide data to clearlyobtain compound-specific adjustment factors to replacedefault or rule of thumb interspecies adjustments. Thework focussed attention on the need to define a dosemetric for interspecies adjustment. Two possibilitiesare peak maternal blood levels or net fetal exposureduring the critical period, which should be proportionalto net area under the boric acid blood curve in maternalplasma.

Criteria Pollutants

Chemicals with extensive history of controlled hu-man experimentation are the criteria pollutants, in-cluding ozone, sulfur dioxide, nitrogen oxides, and car-bon monoxide. These byproducts of commerce and ofthe internal combustion engines that fuel our techni-cal society achieve airborne concentrations quite closeto frank effect levels. Human studies of dosimetry, theamounts retained in the lungs, and pulmonary func-tion in healthy and in compromised individuals havebeen important in setting acceptable ambient levels forthese compounds that strike a balance between safety

and continuing economic activity.

250 DOURSON ET AL.

Organophosphates (OPs)

In adults, at least, the main risks posed from acute ex-posures to OPs are associated with inhibition of tissuecholinesterases and impaired cholinergic transmission.For the thiophosphorate compounds, toxicity is com-plexly related to activation to the active oxon, a pro-cess that has to compete with metabolism, includinghydrolysis to inactive products. How do these variousprocesses work together in target pest species or in in-advertent target species that may be exposed to thesecompounds?

PBPK models have been developed for organophos-phate compounds in order to integrate knowledge ofvarious pathways in both causing toxicity or in inac-tivating the compounds before they can exert their bi-ological effects (Gearhart et al., 1990). The possibilityexists to collect in vitro data with human tissues, blood,etc., to assess kinetic constants for various pathwaysand develop a predictive model for production and dis-tribution of the oxon. The time course of the oxon inblood and tissues would be directly related to inhibitionof the target macromolecules. These predictive modelsshould be validated by limited studies in human vol-unteers. With current analytical methods, these stud-ies can be done at dosages where there are minimalchanges in any of the esterase activities in the body. Themain goal of such work is not simply to assess dosesthat cause some minimal change in blood or plasmacholinesterase. Instead, they fulfill multiple objectivesby assessing the major biological determinants of thekinetics of parent compound and oxon. As with all hu-man studies, doses of these materials have to be care-fully selected and the dosing closely supervised by aphysician. However, human data are critical for makingsound judgements about the use of these compounds, in-suring protection of all exposed individuals, and avoid-ing overreliance of default approaches for animal tohuman extrapolation for such a widely used group ofcompounds.

With this information, it is possible to examine ex-pected differences among individuals associated withmetabolic polymorphisms and to ascertain if the phar-macokinetics seen in other species are expected in hu-mans. In addition, these mechanistic PBPK models canexplore whether there may be specific populations—aged, youth, or compromised individuals—who may beat greater risk. This evaluation is conducted by provid-ing distributions of critical parameters for metabolism,physiological parameters or tissue clearance and seeif particular combinations of parameters give rise topredictions of unusually large tissue doses. The distri-butions entered into the models could include physi-ological and metabolic profiles for children or for theelderly. Often the limiting factor in applying these mod-els with distributions of parameters to estimate tissue

dose/responses is our limited knowledge of parameter

values in the general population or in specific groups inthe general population.

DISCUSSION

Do Human Data Produce More AccurateEstimates of Risk?

This paper focuses on the dose–response assessmentdifferences between humans and experimental ani-mals. At first examination, a significant number of RfDsor RfCs, that we calculate from experimental animaldata and where we use a 10-fold uncertainty factorfor experimental animal to human extrapolation, arehigher than EPA’s human based values. This observa-tion leads us to question whether the 10-fold factor issufficient to protect human health for those animal-based RfDs and RfCs for which we lack human data tocorroborate the nature or dose–response pattern of thecritical effect. The data in Tables 1 and 2 and shown inFig. 1 can be used to address this question, in part. In do-ing so, however, we suggest a more rigorous evaluationof the animal-based RfDs and RfCs that we calculate.This is because our estimations of RfDs or RfCs from ex-perimental animal data have not undergone the samerigor in development and review as those made withthe human data on IRIS (EPA, 2000b).

However, our analysis also show that a significantnumber of RfDs or RfCs, that we calculate from ex-perimental animal data and where we use a 10-folduncertainty factor for experimental animal to humanextrapolation, are lower than the human based values.This observation leads us to question whether, for thoseanimal-based RfDs and RfCs for which we lack hu-man data to corroborate the nature or dose–responsepattern of the critical effect, how many of these RfDsand RfCs err in a direction that overly protects humanhealth? Unfortunately, because our work did not includein-depth and direct comparison of any dose–responsecurves of the critical effect between humans and exper-imental animals because of the general unavailabilityof such data, answers to such questions await additionalanalysis.

Although our analysis indicates that animal data canlead to either a higher or a lower risk value than hu-man data, human data often have provided informationthat reduces uncertainty or identifies a completely dif-ferent end point. For example, it is generally recognizedthat in developing RfDs or RfCs from animal data, theavailability of adequate human data reduces or even ob-viates the need of uncertainty factors for extrapolatingfrom animals to humans. This is based on the presump-tion that such adequate human data are more accuratepredictors of human toxicity than animal data. In addi-tion, EPA (2000b) notes several times when animal dataare deemed inappropriate (e.g., warfarin, manganese,

see Table 4), or the effect is different in at least some

USING HUMAN DATA TO PROTECT THE PUBLIC’S HEALTH 251

TABLE 4Description of Reasons Why an Animal-Based RfD or RfC Is Not Possible or Appropriate

(Data from Tables 1 and 2)

Chemical name(as on EPA’s IRIS) Specie/Type of study Critical effect(s)

Beryllium RfC Human occupational and Beryllium sensitization and progression to chronic beryllium disease (CBD)community exposure

Animal data No laboratory animal model fully mimics all features of human CBDCadmium RfD Human chronic exposures Significant proteinuria

from a variety of studiesAnimal data Insufficient information exists in the IRIS file to make any determination

of an RfDChromium (acid mists Human subchronic Nasal septum atrophy

and aerosols) RfC occupationalAnimal data Experimental animal studies have not reported on nasal mucosal effects

following inhalation exposuresFluorine (soluble fluoride) Human epidemiology Objectionable dental fluorosis

RfD Animal data Insufficient information exists in the IRIS file to make any determination of RfDHydrogen cyanide RfC Human occupational CNS symptoms and thyroid effects

Animal data Insufficient information exists in the IRIS file to make any determination of RfCManganese RfC Human occupation exposure Impairment of neurobehavioral function

Animal data IRIS states that animal toxicity data qualitatively support the human studiesused as a basis of the RfC; quantified comparisons were not provided

Manganese RfD Human data of several types No LOAEL given, CNS effects appear to occur at higher dosesAnimal data IRIS states that human data are superior to any data obtained from animal

toxicity studies as the basis of an RfD, since the physiologicrequirements for manganese vary among species

1,1,2-Trichloro-1,2,2- Human occupational exposure Psychomotor impairmenttrifluoroethane RfD Animal data Insufficient information exists in the IRIS file to make any determination of RfD

Warfarin RfD Human experimental Increased prothrombin timeInappropriate IRIS states “Because of marked differences in the susceptibility of different

species to the effects of warfarin, it would be inappropriate toderive an RfDfrom studies on lower animals”

Zinc and compounds RfD Human experimental diet Decrease in erythrocyte superoxide dismutase concentration in adultssupplement

Animal data Insufficient information exists in the IRIS file to make any determination of RfD

important aspect (e.g., arsenic, barium, beryllium andsilver, see Tables 1 and 2). It is for these reasons, inpart, that EPA gives higher priority to human stud-ies (Barnes and Dourson, 1988; Dourson, 1994; EPA,1994; Jarabek, 1994, 1995). Other organizations havethe same preference in developing their hazard iden-tification and dose–response assessments (Meek et al.,1994; IPCS, 1994).

A recent text by Olson et al. (2000) also focuses onthe hazard identification comparisons of experimentalanimals and humans for a series of 150 pharmaceu-tical compounds. These authors conclude that concor-dance between human and experimental animal toxic-ity was highest in the hematological, gastrointestinal,and cardiovascular areas and poorest for cutaneous, en-docrine, and hepatobiliary and liver function abnormal-ities. Overall for 30% of the human toxicities, there wasno relationship with toxicities seen in animals. Thiswork supports the idea that in a significant number oftimes, human data would be essential for hazard iden-tification and presumably for dose–response assess-

ment.

Every cancer that has been first associated with achemical in human studies, for which numerous exam-ples exist (e.g., benzene, asbestos, chromium), supportsthe idea that human data result in a lower “safe” dosewhen compared to animal data. Animal bioassays maynow be able to better predict carcinogenicity in humansthan in the days prior to mandatory testing and the de-velopment of test guidelines, but other uncertainties aremore likely to exist. The epidemiological data on dioxinsuggest that humans are far less sensitive than animals(Hays et al., 1997). In contrast, Crump et al. (1989) showrough similarities among human or animal studies for23 chemicals for which adequate data were available toestimate lifetime cancer doses associated with a 25%incidence of tumor response (i.e., TD25s) on the basis ofmg/kg body weight dose. However, there is no reasonto believe that for some other chemical the error in anassessment based on animal data would be the otherway, with humans being far more sensitive (for exam-ple, see the results in Table 1 for 2,4-dinitrophenol). Thequestion for either the cancer or noncancer end point

is the same: if we ignore opportunities for collecting

252 DOURSON ET AL.

human data to check our assessments, will we be bas-ing standards on the wrong critical effect, or the wrongassumptions and consequently setting limits too highto be protective of the public’s health?

A recommendation that might follow from these ob-servations is that an evaluation of data from humans,when available and judged to be sufficient, is essentialto the development of RfDs and RfCs. Such data seempreferable to using a uncertainty factor of 10 for ex-perimental animal to human extrapolation, and oftenidentify effects not seen in animal studies.

What Are Some of the Limitations ofOur RfD/RfC Analysis?

The ratios of human to animal based RfDs and RfCsfound in Tables 1 and 2 and in Fig. 1 should only be con-sidered as a first approximation of the value of humandata in the determination of a RfD or RfC for protectionof the public’s health. This is because the estimation ofthe RfD or RfC based on the human study found onEPA’s IRIS (or elsewhere) was from:

• A thorough analysis of the available data based ona review of original studies,• The development of a risk assessment document,

and• Debate in one or more internal ATSDR, EPA, or

Health Canada peer review meetings, and, in at leastthe case of EPA, unanimous acceptance.

By contrast, the animal-based RfDs and RfCs that wedevelop here were generally based on summary infor-mation primarily found on EPA’s IRIS or TERA’s ITER.These animal-based RfDs and RfCs generally did nothave the benefit of the development of a risk document,nor extensive peer review. A more comprehensive anal-ysis of animal-based RfDs and RfCs would have meant acomparable level of analysis and peer review, but clearlya much more intensive involvement, than this paperallows.

We have made these comparisons between humanand animal data on the basis of the RfD or RfC. In otherwords, the comparisons were made after uncertaintyfactors have been applied to the NOAEL, LOAEL,BMD, or BMC. This approach is consistent with ourgoal, which is to evaluate how well human data, usedwith contemporary risk assessment practices, protecthuman health. The range of uncertainty factors thatis used with the NOAELs, LOAELs, BMD, or BMCin these tables reflects the variety of data types anddatabases for different chemicals. Therefore, we basedour comparison on the RfDs and RfCs themselves, noton the NOAELs, etc. However, we acknowledge that itwould be perhaps better if comparisons could be madeon specific matching end points between the experimen-tal animal species and humans, such as what has re-cently been done by EPA for the reassessment of dioxin

(http://www.epa.gov/ncea/pdfs/dioxin/dioxreass.htm).

If high-quality animal data are available, the defaultuncertainty factor is likely to be 100 for RfDs (e.g., ni-trite, malathion) and a 10-fold difference between an-imal and average human NOAEL may be expected.For the chemicals with human and animal RfDs basedon the same biological end point and data type (e.g.,NOAELs), a ratio close to 1 corroborates the selection ofan interspecies uncertainty factor of 10-fold. For exam-ple, for aldicarb (i.e., plasma cholinesterase inhibition)and baygon, the ratios are similar only with an uncer-tainty factor of 10-fold. A body of ongoing research seeksto evaluate and improve the basis for the uncertaintyfactors (e.g., Dourson et al., 1996; Renwick and Lazarus,1998; Meek et al., 2000).

This issue of power of a study to detect change is alsoimportant, but generalizations about the power of dif-ferent studies, such as the idea than animal studiesare always more powerful than human studies becausethey often have more subjects, are difficult. We suggesta case by case comparison—a large case control studymay have a few hundred people; a cohort study mayhave thousands, some cohorts follow up the people un-til nearly all have died, but some epidemiology studieshave only limited exposure data.

A major issue here is the power of any study, but inparticular, the power of a “negative” epidemiology study.In cancer risk assessment negative epidemiology stud-ies have been used to set upper limits on the possiblerisk—if a study had the power to detect a 3-fold increaseand does not, it assures us that risks above 3X are un-likely. Issues of power include not only study size, butalso design and exposure assessment.

We defined several criteria for comparing the exist-ing human-based RfDs and RfCs with the animal-basedRfDs and RfCs we estimate in our paper. The lowestcriterion was the comparison of RfDs and RfCs basedon NOAELs for unrelated effects. This is, of course, theleast preferred comparison because the human and ani-mal data tell us different things about the toxicity of thechemical in question. We made this choice quite oftenin Tables 1 and 2.

The choice between a NOAEL or BMD (or their cor-responding inhalation counterparts) for the same or re-lated critical effect were the next best criteria in thecomparison of human and animal-based RfDs and RfCs.Of course, these are more preferred comparisons be-cause the human and animal data tell us more simi-lar things about the toxicity of the chemical in ques-tion. We also made this choice quite often in Tables 1and 2.

Some might suggest that such comparisons should bemade on the basis of the BMD rather than the NOAEL.We have no inherent problem with this suggestion (ourchoices here were based on availability of BMDs). How-ever, in general, the choice of either a NOAEL or BMDas the basis of this comparison correctly depends on theavailable data. For example, dog studies seldom have a

sufficient number of animals to do an adequate BMD,

USING HUMAN DATA TO PROTECT THE PUBLIC’S HEALTH 253

but the value in exploring different types of effects witha larger animal should not be underestimated. NOAELsfrom dogs might be vastly superior to a BMD from ro-dents, if blood pressure was the critical effect, for ex-ample. In addition, not all effects model well. Numer-ous examples exist of critical effects that are masked athigher doses by effects of greater severity. The BMDcan often not adequately model these critical effectsand risk assessors need to depend on the NOAEL orLOAEL for guidance. Risk assessors often analyze theeffects of different severity at every dose of a chemical,and recall their experiences in evaluating the resultsof other experiments with similar chemicals, beforethey make judgments of critical effect. Such judgementcannot depend only on the estimation of BMDs, norNOAELs.4

Unfortunately, we did not have sufficient data in or-der to match any dose–response curves of the criticaleffect between human- and experimental animal-basedRfDs and RfCs. Matching such curves is recognized asbeing an almost ideal criterion, but because many dataare needed for such comparison, it is also the least likelycriterion to be fulfilled.

Do Alternatives to Human Clinical Designs Existin Reference to Pharmacokinetics?

The goal of standard setting is to establish exposurelevels that are protective of human health in a va-riety of situations—environmental exposures, occupa-tional exposures, and exposures to by-products of ourmodern society. The intellectual tools for all these ex-ercises are similar. These exercises require interpreta-tion of a body of data on effects in animals or in ex-posed individuals to assess minimally toxic levels andto apply specific factors to insure that all members ofexposed populations receive adequate protection. Theperceived safe level may vary for criteria pollutants,essential nutrients, and occupational toxicants. How-ever, in all cases, considerations of similar factors be-come important. Pharmacokinetic models or tools aredesigned to determine conditions where similar doses totissues would be attained for different species or expo-sure situations. These affect the numerator in the RfDor RfC process. PBPK modeling evaluates critical deter-minants for the pharmacokinetics and aids in assessinginterindividual variability and identifying at-risk popu-lations. These considerations affect the denominator inthe RfD or RfD determination. These PK tools togetherwith human pharmacokinetic data from volunteerstudies and in vitro mechanistic studies with humantissues provide an optimal data set from which sounddecisions can be reached for protection of exposed indi-

viduals.

4 This is why the BMD and NOAEL need to be somewhat compara-ble; if they are not comparable, then the risk assessors’ job of criticaleffect judgment becomes exceedingly difficult with data sets havingboth BMD and NOAEL.

PBPK approaches are important because they use sci-entific methods to as accurately as possible estimateand understand potential risk to humans and providegreater scientific certainty in risk characterization. Thealternatives to PBPK are default assumptions, whichhave greater uncertainty.

If Human Data Are the Preferred Basis to ProtectHuman Health, Then What Does One Conclude ifHuman Data Lead to a Higher Risk Value ThanOne Based on Animal Data?

All ratios in Tables 1 and 2 that have a value greaterthan 1 demonstrate that the human based RfD or RfCis greater than the animal-based value. Ratios aboutequal to 10 would be expected if animals and humanswere shown to be about equally sensitive, that is ifNOAELs or LOAELs were similar (e.g., see chlorpyri-fos or ethephon in Table 1). This is because the use ofa 10-fold interspecies uncertainty factor with the an-imal NOAEL or LOAEL would result in a RfD or RfCthat was about 10 times lower than a human-based RfD(and thus the ratio of human to animal-based risk valuewould be greater than 1). Even if the NOAEL in hu-mans were lower than in animals, but by a factor of∼10 or less, the use of an interspecies uncertainty fac-tor of 10-fold could result in a lower RfD or RfC basedon animal data than the RfD or RfC based on humandata.

Such a situation would also occur if the human datawere suggesting a critical effect that the animal datawere not. Table 1 shows several examples of this (e.g.,nitrate, nitrite and selenium and compounds). In somecases, EPA and others judge that the animal data arenot a reliable basis of the RfD or RfC and a ready com-parison is not available or recommended (e.g., man-ganese, warfarin). This is an example of the importantrole of data other than whole animal bioassays, such aspharmacokinetics and pharmacodynamics, in the over-all determination of RfDs and RfCs.

What is more problematic, and potentially much moreserious, is when the human data suggest a lower RfDor RfC than the animal data, either because of greatersensitivity to the critical effect, or different critical ef-fect. This occurs in 36% (12 of 33 comparisons) of thecomparisons we performed, if no precision issues areconsidered, and occurs in 15% (5 of 33 comparisons) ifprecision is considered to be ”perhaps an order of magni-tude.” In either case, the use of animal-based RfDs andRfCs, rather than those based on human data mightbe considered inappropriate because the public’s healthdoes not appear to be protected.

If we accept the premise that human data are morereliable and relevant for a human risk assessment, thenit follows that human data should be used without biasas to whether their use results in a higher or lower RfDor RfC. Replacement of animal data with human data

should be dependent on the quality of the human data

254 DOURSON ET AL.

being at a minimum comparable to the animal data itwill replace. Focus should be placed on a comparisonof the quality of the human data to the animal dataand the relevance and uncertainty for human health ofeach.

CONCLUSIONS AND FUTURE DIRECTIONS

At the outset we asked a series of questions regardingthe use of human data for risk assessment.

• Is information from human studies the best way tojudge the potential health risk from chemicals in ourenvironment?

The answer is perhaps given best by the many healthorganizations that address this question. For exam-ple, Health Canada (Meek et al., 1994), IPCS (1994),and EPA (Barnes and Dourson, 1988; EPA, 1994) eachprefers the use of human data rather than animal datain the development of risk values. Based on our analy-sis we also agree. For example, the use of human datahas led to the estimation of RfDs and RfCs on EPA’sIRIS that are often lower than animal-based RfDs andRfCs that we estimate in this paper. Especially in thesecases, the use of these human-based RfDs and RfCs maybe better protective of the public’s health.• Should public health agencies strive to resolve the

ethical questions and identify criteria to foster the con-duct of human studies that might lead to better protec-tion of the public’s health?

We hope that the answer to this question is a resound-ing yes. Our analysis, although limited, suggests a highpercentage of comparisons where the exclusive use ofanimal data would lead to RfDs and RfCs that do notprotect the public’s health. Moreover, in many cases theanimal data were recognized as having greater limita-tions than the existing human data, as reflected in gen-erally larger uncertainty factors than those used withthe existing human data.

Perhaps what we really should be asking is how goodare our best-qualified animal data for protecting humanhealth? How can we determine when it would be betterto use or develop human data rather than conductinganother 2- year animal study?• Should scientists ignore available human data that

might suggest a lower or higher risk value?

The obvious answer here is no. Protecting the public’shealth is why we all do this work. However, it seems rea-sonable for scientists and regulatory agencies to furtherevaluate the use of human data in the development ofrisk values, such as RfDs and RfCs. Perhaps new cri-teria could be developed so that such data can be used

with greater confidence.

Additional analysis of the results we present herecould be done. For example, we were able to use thecomplete listing of RfDs and RfCs based on human datafrom EPA’s IRIS. Other organizations also have simi-lar risk values based on human data, and these valuesmight also be compared with those based on experimen-tal animal data. In addition, further research might ex-plore whether commonalties exist among target tissueor mode of action for those chemicals for which animal-and human-based RfDs and RfCs differ quantitatively.None of the animal-based RfCs that we estimated wasbelow the human-based one found on IRIS, within thelimits of precision being assumed. Was this deficiencydue to a small sample size or was it related to the useof dosimetry for RfCs, but not yet for RfDs? Similarly,what are the implications for the health protective-ness of the vast majority of the RfDs and RfCs basedon animal data, without any human data to act as acheck?

We look toward future collaborations with other in-terested scientists for resolution of these and otherissues.

ACKNOWLEDGMENTS

We thank Lynne Haber of Toxicology Excellence for Risk Assess-ment, Ruthann Rudel of Silent Spring Institute, Russ Savage of theNational Institute for Occupational Safety and Health, and BernSchwetz of the Food and Drug Administration for insightful com-ments. We also thank Meg Poehlmann for editing and administra-tive assistance. Support for this research was provided through theuse of developmental reserve funds of Toxicology Excellence for RiskAssessment (TERA), Cincinnati, Ohio, a 501(c) (3) nonprofit organi-zation.

REFERENCES

Andersen, M. E., and Jarabek, A. M. (2000). Nasal tissue dosimetry—Issues and approaches for “Category I” gases: A report on a meetingheld in Research Triangle Park, NC, February 11–12, 1998. Inhal.Toxicol., in press.

Andersen, M. E., Sarangapani, R., Frederick, C. B., and Kimbell,J. S. (1999). Dosimetric adjustment factors for methylmethacry-late derived from a steady-state analysis of a physiologically basedclearance-extraction model. Inhal. Toxicol. 11, 899–926.

Andersen, M. E., Clewell, H. J., III, Gargas, M. L., MacNaughton,M. G., Reitz, R. H., Nolan, R., and McKenna, M. (1991). Physio-logically based pharmacokinetic modeling with dichloromethane,its metabolite, carbon monoxide, and blood carboxyhemoglobinin rats and humans. Toxicol. Appl. Pharmacol. 108, 14–27.

Andersen, M. E., Clewell, H. J., III, Gargas, M. L., Smith, F. A., andReitz, R. H. (1987). Physiologically-based pharmacokinetics and therisk assessment process for methylene chloride. Toxicol. Appl. Phar-macol. 87, 185–205.

Barnes, D. G., and Dourson, M. L. (1988). Reference dose (RfD): De-scription and use in health risk assessments. Regul. Toxicol. Phar-macol. 8, 471–486.

Barton, H. A., Andersen, M. E., and Clewell, H. J., III (1998). Har-monization: Developing consistent guidelines for applying mode of

USING HUMAN DATA TO PROTECT THE PUBLIC’S HEALTH 255

action and dosimetry information to cancer and noncancer risk as-sessment. Hum. Ecol. Risk Assess. 4, 75–115.

Clewell, H. J. (1995). Incorporating biological information in quanti-tative risk assessment: An example with methylene chloride. Tox-icology 102, 83–94.

Crump, K., Allen, B., and Shipp, A. (1989). Choice of dose mea-sure for extrapolating carcinogenic risk from animals to humans:An empirical investigation of 23 chemicals. Health Phys. 57, 387–393.

DeGeorge, J. (1999). Transcript of the USEPA Joint Science Advi-sory Board/Science Advisory Panel, Data from Testing on HumanSubjects Subcommittee, November 30, 1999.

Dourson, M. L. (1994). Methods for establishing oral reference doses(RfDs). In Risk Assessment of Essential Elements. (W. Mertz, C. O.Abernathy, and S. S. Olin, Eds.), pp. 51–61. ILSI Press, Washington,DC.

Dourson, M. L., Felter, S. P., and Robinson, D. (1996). Evolution ofscience-based uncertainty factors in noncancer risk assessment.Regul. Toxicol. Pharmacol. 24, 108–120.

Dourson, M. L., Meier, A., Meek, B., Renwick, A., Ohanian, E., andPoirier, K. (1998). Boron tolerable intake: Re-evaluation of toxicoki-netics for data-derived uncertainty factors. Biol. Trace Elem. Res.66(1–3), 453–463.

Elwood, J. M. (1998). Critical Appraisal of Epidemiological Studiesand Clinical Trials, Oxford Univ. Press, Oxford.

EPA (Environmental Protection Agency) (1994). Methods for Deriva-tion of Inhalation Reference Concentrations and Application of In-halation Dosimetry, EPA/600/8-90-066F, October. Office of Healthand Environmental Assessment, Washington, DC.

EPA (Environmental Protection Agency) (1999). The Office Of Pes-ticide Programs’ Policy on Determination of the Appropriate FQPASafety Factor(s) for Use in the Tolerance-Setting Process. May. Officeof Pesticide Programs.

EPA (Environmental Protection Agency) (2000a). Science AdvisoryBoard. Executive Committee Review Draft Report of the JointSAB/SAP Subcommittee on Data from Testing of Human Subjects.Available at http://www.epa.gov/sab/Reports/2000report/2000dra-fts/Humandata. May 31, draft.

EPA (Environmental Protection Agency) (2000b). Integrated Risk In-formation System (IRIS). National Center for Environmental As-sessment. See listing for individual chemicals and the glossaryfor risk assessment terms. Available online at http://www.epa.gov/iris.

Felter, S. F., and Dourson, M. L. (1998). The inexact science of riskassessment (and implications for risk management). Human Ecol.Risk Assess 2, 245–251.

Frederick, C., Bush, M. L., Subramaniam, R. P., Black, K. A., Finch,L., Kimbell, J. S., Morgan, K. T., Morris, J. B., and Ultman, J. S.(1998). Application of a hybrid computational fluid dynamics andphysiologically based inhalation model for interspecies dosimetryextrapolation of acidic vapors in the upper airways. Toxicol. Appl.Pharmacol. 152, 211–231.

FDA (Food and Drug Administration) (1997). FDA Public Health Ad-visory, Reports of Valvular Heath Disease in Patients Receiving Con-comitant Fenfluramine and Phentermine. July.

Gearhart, J. M., Jepson, G. W., Clewell, H. J., III, Andersen, M. E.,and Conolly, R. B. (1990). A physiologically based model for in vivoinhibition of acetylcholinesterase by diisopropyl fluorophosphate.Toxicol. Appl. Pharmacol. 106, 295–310.

Gerlowski, L. E., and Jain, R. K. (1983). Physiologically based phar-macokinetic modeling: Principles and applications. J. Pharm. Sci.72, 103–1127.

Hays, S., Aylward, L., Karch, N., and Paustenbach, D. (1997). The

relative susceptibility of animals and humans to the carcinogenic

hazard posed by exposure to 2,3,7,8-TCDD: An analysis using stan-dard and internal measures of dose. Chemosphere 34(5–7), 1507–1522.

ILSI (International Life Sciences Institute) (1997). An evaluation ofEPA’s proposed guidelines for carcinogen risk assessment usingchloroform and dichloroacetate as case studies: Report of an expertpanel. November. Washington, D.C.

IPCS (International Programme on Chemical Safety) (1994). Deriva-tion of guidance values for health-based exposure limits. Environ-mental Health Criteria No. 170: Assessing Human Health Risks ofChemicals. World Health Organization, Geneva.

Jarabek, A. M. (1994). Inhalation RfC methodology: Dosimetricadjustments and dose-response estimation of noncancer toxicityin the upper respiratory tract. Inhal. Toxicol. 6(Suppl.), 301–325.

Jarabek, A. M. (1995). The application of dosimetry models to iden-tify key processes and parameters for default dose–response as-sessment approaches. Toxicol. Lett. 79, 171–184.

Lee, ET. (1980). Statistical Methods for Survival Data Analysis. Life-time Learning Publications, CA.

MacGregor, J. T., Collins, J. M., Sugiyama, Y., Tyson, C. A., Dean, J.,Smith, L., Andersen, M., Curren, R., Houston, J. B., Kadlubar, F. K.,Kedderis, G. L., Krishnan, K., Li, A., Parchment, R. E., Thummel,K., Tomaszewski, J. E., Ulrich, R., Vickers, A. E. M., and Wrighton,S. A. (2000). In vitro Human tissue models in risk assessment: Re-port of a consensus-building workshop In vitro human tissue mod-els in risk assessment. Toxicol. Sci., in press.

Meek, M. E., Newhook, R., Liteplo, R. G., and V. C. Armstrong. (1994).Approach to assessment of risk to human health for priority sub-stances under the Canadian Environmental Protection Act. Envi-ron. Carcinogen. Ecotoxicol. Rev. C12(2), 105–134.

Meek, B., Renwick, A., Ohanian, E., Dourson, M., Lake, B., Naumann,B., and Vu, V. (2001). Guidelines for application of compound spe-cific adjustment factors (CSAF) in dose/concentration response as-sessment. In press.

Murray, F. J., and Andersen, M. E. (2000). Data-derived uncertaintyfactors: Boric acid (BA) as a case study. Hum. Ecol. Risk Assess., inpress.

NAS (National Academy of Sciences) (1983). Risk Assessment in theFederal Government: Managing the Process. National AcademyPress, Washington, DC.

NAS (National Academy of Sciences) (1994). Science and Judg-ment in Risk Assessment, National Academy Press, Washington,DC.

NCEA (National Center for Environmental Assessment) (2000).Exposure and Human Health Reassessment of 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and Related Compounds.Online at http://www.epa.gov/ncea/pdfs/dioxin/dioxreass.htm.

NRC (National Research Council) (1986). Dose-Route Extrapolations:Using Inhalation Toxicity Data to Set Drinking Water Limits.Chapter 6, Drinking Water and Health, Volume 6: NationalAcademy Press, Washington, DC.

Okoji, R., Leininger, J., and Froines, J. (1999). Subchronic toxicitystudy of sodium arsenite in methyl-deficient male C57BL/6 mice.In: Arsenic Exposure and Health Effects (W. Chappell, C. Abernathy,and R. Calderon, Eds.), pp. 225–232. Elsevier, Amsterdam, TheNetherland.

Olson, H., Betton, G., Robinson, D., et al. (2000). Concordance of thetoxicity of pharmaceuticals in humans and in animals. Regul. Tox-icol. Pharmacol. 32, 56–67.

Plowchalk, D. R., Andersen, M. E., and Bogdanffy, M. S. (1997).Physiologically-based modeling of vinyl acetate uptake, metabolismand intracellular pH changes in the rat nasal cavity. Toxicol. Appl.

Pharmacol. 142, 386–400.

256 DOURSON ET AL.

Reitz, R. H., Mendrela, A. L., and Guengerich, F. P. (1989).In vitro metabolism of methylene chloride in humanand animal tissues: Use in physiologically based phar-macokinetic models. Toxicol. Appl. Pharmacol. 97, 230–246.

Renwick, A. G. (1993). Data derived safety factors for the evaluation offood additives and environmental contaminants. Food Addit. Con-tam. 10(3), 275–305.

Renwick, A., and Lazarus, N. R. (1998). Human variability and non-cancer risk assessment–An analysis of the default uncertainty fac-tor. Regul. Toxicol. Pharmacol. 27, 3–20.

Russo, E. (2000). Monitoring human subjects and clinical trials. Sci-

entist May 15, p. 6.

SOT (Society of Toxicology) (2000). The Value and Ethics of UsingHuman Data for the Registration of Pesticides. 39th Annual Meet-ing, March 23.

TERA (Toxicology Excellence for Risk Assessment) (2000). Inter-national Toxicity Estimates for Risk (ITER) Database. Online atwww.tera.org.

Thomas, R. S., Bigelow, P. L., Keefe, T. J., and Yang, R. S. H.(1996). Variability in biological exposure indices using physiologi-cally based pharmacokinetic modeling and Monte Carlo simulation.Amer. Ind. Hyg. Assoc. J. 57, 23–32.

Vaziri, N. D., and Oveisi, F. (2000). Evaluation of Renal Clearance ofBoric Acid in Non-pregnant and Pregnant Sprague–Dawley Rats.

Final Report. Univ. California, Irvine, April.