DRAFT                                                 EPA/100/M1/001
DO NOT CITE OR QUOTE                                  External Review Draft
                                                       May 2011
           Guidance for Applying Quantitative Data
      to Develop Data-Derived Extrapolation Factors for
          Interspecies and Intraspecies Extrapolation
                                NOTICE

THIS DOCUMENT IS AN EXTERNAL REVIEW DRAFT. It has not been formally released
by the U.S. Environmental Protection Agency and should not at this stage be construed to
represent Agency policy. It is being circulated for comment on its technical accuracy and policy
implications.
                         Office of the Science Advisor
                           Risk Assessment Forum
                     U.S. Environmental Protection Agency
                           Washington, DC 20460

-------
                                     DISCLAIMER

       This document is distributed solely for the purpose of predissemination peer review under
applicable information quality guidelines. It has not been formally disseminated by EPA.  It
does not represent and should not be construed to represent any Agency determination or policy.
Mention of trade names or commercial products does not constitute endorsement or
recommendation for use.

                                      ABSTRACT

       This document describes the U.S. EPA's approach for developing quantitative factors for
extrapolating effect levels from animals to humans and to address human variability. These
extrapolations have been covered by the uncertainty factors UFA and UFn, respectively. In the
absence of quantitatively-valuable data, default values for these uncertainty factor values may be
applied. However, informative data that describe variability in chemical distribution
(toxicokinetics, TK) and dose-response (toxicodynamics, TD) should be first considered. This
document describes the separation of UFA and UFn into TK and TD components and describes
the process for identifying pertinent data useful for quantifying inter- and intraspecies differences
to serve as the basis for nondefault, data-derived extrapolation factors (DDEFs). Key
considerations include identifying a tissue concentration associated with a given response level,
and identifying and measuring a biological response associated with the corresponding toxicity.
Interspecies TK variability is quantified on the basis of doses or concentrations that produce the
same tissue concentration in animals and humans; intraspecies TK variability is defined as
differences in tissue concentration attained from the same human exposure; TD variability is
defined on the basis of doses or concentrations that produce the same response. This approach is
consistent with the approach for deriving reference concentration (RfC) values; it represents a
point in the continuum of approaches that includes default approaches, categorical default values
(e.g., body-weight scaling), and integrated biologically based dose-response models.
           This document is a draft for review purposes only and does not constitute Agency policy.
5/11/11                                      ii         DRAFT: DO NOT CITE OR QUOTE

-------
                                   CONTENTS
LIST OF TABLES	v
LIST OF FIGURES	v
LIST OF ABBREVIATIONS	vi
PREFACE	viii
AUTHORS, CONTRIBUTORS AND REVIEWERS	ix

1.  INTRODUCTION	1
   1.1.  BACKGROUND	1
   1.2.  PURPOSE AND SCOPE	2

2.  TECHNICAL CONCEPTS AND PRINCIPLES FOR DDEF	5
   2.1.  BACKGROUND	5
   2.2.  EXTRAPOLATION WITH DDEFs	7
       2.2.1. Approaches to Deriving DDEFs	7
             2.2.1.1.  TK and TD Models	8
             2.2.1.2.  Use of Ratios to Calculate DDEF	10
       2.2.2. Qualitative Considerations	13
       2.2.3. Information Quality	13
       2.2.4. Additional Considerations	14
             2.2.4.1.  Mode of Action	14
             2.2.4.2.  Use of In Vitro Data	15
             2.2.4.3.  Uncertainty and Variability	16
             2.2.4.4.  Multiple Critical Effects	17
             2.2.4.5.  Screening Level vs. Refined Risk Assessments	17

3.  DDEFs BASED ON TOXICOKINETICS (TK)	18
   3.1.  GENERAL CONSIDERATIONS	18
       3.1.1. Dose Metric	19
       3.1.2. Dose Selection	22
       3.1.3. In Vitro Data	22
   3.2.  INTERSPECIES TOXICOKINETIC EXTRAPOLATION (EFAK)	23
       3.2.1. Considerations for £FAK	23
       3.2.2. Computation	25
             3.2.2.1.  Use of TK Models	25
             3.2.2.2.  Use of Ratios	25
       3.2.3. Relationship to Other EPA Guidance	27
       3.2.4. Conclusions for EFAK	28
   3.3.  INTRASPECIES TOXICOKINETIC EXTRAPOLATION (EFHK)	29
       3.3.1. Considerations for EFnK	29
             3.3.1.1.  Susceptible Groups or Individuals	29
             3.3.1.2.  Target Tissues	31
             3.3.1.3.  Dose Response	31

          This document is a draft for review purposes only and does not constitute Agency policy.
5/11/11                                  iii       DRAFT: DO NOT CITE OR QUOTE

-------
                             CONTENTS (continued)
       3.3.2. Computation	32
             3.3.2.1. Use of TK Models	32
             3.3.2.2. Use of Ratios	32
       3.3.3. Conclusions forEFnK	33

4.  DDEFs BASED ON TOXICODYNAMICS (TD)	34
   4.1.  GENERAL CONSIDERATIONS	34
       4.1.1. Mode of Action	35
       4.1.2. Relating Response to Dose or Concentration	35
       4.1.3. Range of Doses or Concentrations	36
   4.2.  INTERSPECIES TOXICODYNAMIC EXTRAPOLATION (EFAD)	36
       4.2.1. Considerations for £FAD	37
             4.2.1.1. Mode of Action	37
             4.2.1.2. Target Tissues	37
             4.2.1.3. Dose Response	38
             4.2.1.4. In Vitro Data	38
       4.2.2. Computation	39
             4.2.2.1. Use of TD Models	39
             4.2.2.2. Use of Ratios	39
       4.2.3. Conclusions for EFAD	40
   4.3.  INTRASPECIES TOXICODYNAMIC EXTRAPOLATION (EFHD)	40
       4.3.1. Considerations for EFno	41
             4.3.1.1. Susceptible Groups or Individuals	41
             4.3.1.2. Target Tissues	41
             4.3.1.3. In Vitro Data	41
       4.3.2. Computation	42
             4.3.2.1. Use of TD Models	42
             4.3.2.2. Use of Ratios	42
       4.3.3. Conclusions forEFno	44

5.  FINAL STEPS	46

6.  REFERENCES	47

APPENDIX: CASE STUDIES TO ACCOMPANY DATA-DERIVED
            EXTRAPOLATION FACTOR DOCUMENT	A-l
          This document is a draft for review purposes only and does not constitute Agency policy.
5/11/11                                   iv       DRAFT: DO NOT CITE OR QUOTE

-------
                                   LIST OF TABLES

Table 1. Example equations used to derive DDEFs	11

                                  LIST OF FIGURES

Figure 1. Derivation of RfDs/RfCs using uncertainty factors	6
Figure 2. Decision process for DDEFs	9
Figure 3. Interspecies toxicokinetics	24
Figure 4. Interspecies extrapolation based on AUC	27
Figure 5. Intraspecies toxicokinetics	30
Figure 6. Interspecies toxicodynamics	37
Figure 7. Intraspecies toxicodynamics	43
           This document is a draft for review purposes only and does not constitute Agency policy.
5/11/11                                     v        DRAFT: DO NOT CITE OR QUOTE

-------
                             LIST OF ABBREVIATIONS
ADME
AUC
AUCsens
AUCA
AUCgen

AWQCD
BBDR
BMD
BMDL
BMDL05
Cl
Clint
^max
CSAFs
CYP
DDEFs
EGBE
HED
ILSI
IPCS
IRIS
Km
LOAEL
MOA
NOAEL
NRC
PBPK
POD
RfC
RfD
TD
TK
U.S. EPA
UF
UFA
EFAK
UFH
   absorption, distribution, metabolism, elimination; toxicokinetics
   area under the curve
   area under the curve at a percentile of interest in the sensitive human population
   area under the curve in the animal
   area under the curve at a measure of central tendency in the general human
   population
   Ambient Water Quality Criterion document
   biologically based dose response
   benchmark dose
   benchmark dose corresponding to a 10% increase in response
   lower 95% confidence limit of the benchmark dose
   benchmark dose corresponding to a 5% increase in response
   clearance
   intrinsic clearance
   maximum concentration
   chemical-specific adjustment factors
   cytochrome P450
   data-derived extrapolation factors
   effective dose producing a 10% response rate
   ethylene glycol monobutyl ether
   human equivalent dose
   International Life Sciences Institute
   International Programme on Chemical Safety
   Integrated Risk Information System
   Michaelis constant, substrate concentration at rate of 1/2 Vmax
   lowest-observed-adverse-effect level
   mode of action
   no-observed-adverse-effect level
   National Research Council
   physiologically-based pharmacokinetic
   point of departure
   reference concentration
   reference dose
   toxicodynamics
   toxicokinetics
   U.S. Environmental Protection Agency
   uncertainty factor
   interspecies uncertainty factor
   toxicodynamic component of interspecies extrapolation factor
   toxicokinetic component of interspecies extrapolation factor
   intraspecies uncertainty factor
5/11/11
This document is a draft for review purposes only and does not constitute Agency policy.
                                 vi        DRAFT: DO NOT CITE OR QUOTE

-------
                        LIST OF ABBREVIATIONS (continued)
              toxicodynamic component of intraspecies extrapolation factor
EFHK         toxicokinetic component of intraspecies extrapolation factor
Vmax          theoretical maximal initial velocity
WHO         World Health Organization
           This document is a draft for review purposes only and does not constitute Agency policy.
5/11/11                                     vii        DRAFT: DO NOT CITE OR QUOTE

-------
                                       PREFACE

       This guidance document is intended to help U.S. Environmental Protection Agency
(EPA) risk assessors take a data-based approach to nonlinear low-dose extrapolation.  It
describes data recommendations and methods or procedures to calculate data-derived
extrapolation factors (DDEFs) for chemicals for inter- and intraspecies extrapolation.  While
serving the same purpose as uncertainty factors, these extrapolation factors are based on data
directly pertinient to the chemical of interst, rather than having their basis on default assumptions
about inter- and intra-species variability. EPA's 2005 Guidelines for Carcinogen Risk
Assessment (or "Cancer Guidelines'") (U.S. EPA, 2005) describes the EPA's current practice of
examining all relevant and available data first when performing a risk assessment. When
chemical—and/or—site-specific data are unavailable or insufficient, the EPA uses default
assumptions or processes in order to continue with the risk assessment.  Under this practice, EPA
guidance suggests invoking defaults only after the data are determined not to be usable at a
particular point in the assessment.  Both the Cancer Guidelines and the EPA document, An
Examination of EPA Risk Assessment Principles and Practices: a Staff Paper (U.S. EPA, 2004),
note that this is a shift in paradigm from the older practice of assuming that default values or
processes will be used unless there are sufficient data to warrant a departure from the default.
Neither document describes specific approaches to using data rather than default uncertainty
factors in low-dose extrapolation.  This document, Guidance for Applying Quantitative Data to
Develop Data-Derived Extrapolation Factors for Inter species and Intraspecies Extrapolation,
describes those approaches and processes.
       The members of the Risk Assessment Forum Technical Panel on Data-Derived
Extrapolation Factors emphasize that the information offered here is neither a checklist nor a set
of requirements that must be met in order to perform inter- and intraspecies dose extrapolation.
It presents guidance and codifies many of the best practices available when evaluating data for
the purpose of inter- and intraspecies extrapolation factor development. This will contribute to
consistency in the EPA processes and decisions.
           This document is a draft for review purposes only and does not constitute Agency policy.
5/11/11                                      viii       DRAFT: DO NOT CITE OR QUOTE

-------
                  AUTHORS, CONTRIBUTORS, AND REVIEWERS
AUTHORS
John Lipscomb, Co-Lead, U.S. Environmental Protection Agency, Office of Research and
       Development, Cincinnati, OH 45268
Anna Lowit, Co-Lead, U.S. Environmental Protection Agency, Office of Pesticide Programs,
       Arlington, VA 22202
Brenda Foos, U.S. Environmental Protection Agency, Office of Children's Health Protection,
       Washington, DC 20460
Audrey Galizia, U.S. Environmental Protection Agency, Office of Research and Development,
       Edison, NJ 08837
Elaina Kenyon, U.S.  Environmental Protection Agency, Office of Research and Development,
       RTF, NC 27709
Ginger Moser,  U.S. Environmental Protection Agency, Office of Research and Development,
       RTF, NC 27709
Rita Schoeny, U.S. Environmental Protection Agency, Office of Water, Washington, DC 20460
CONTRIBUTORS
Rory Conolly, U.S. Environmental Protection Agency, Office of Research and Development,
       RTF, NC 27709
Deirdre Murphy, U.S. Environmental Protection Agency, Office of Air and Radiation, RTF, NC
       27709
Bruce Rodan, U.S. Environmental Protection Agency, Office of Research and Development,
       Washington, DC 20460
Risk Assessment Forum Staff
Michael Broder, Science Coordinator/DDEF Project Staff Lead, U.S. Environmental Protection
       Agency, Office of the Science Advisor, Washington, DC 20460
Gary Bangs, Acting Executive Director, 2009, U.S. Environmental Protection Agency, Office of
       the Science Advisor, Washington, DC 20460
Kathryn Gallagher, Executive Director, 2010, U.S. Environmental Protection Agency,
       Washington, DC 20460
Elizabeth Lee Hofmann, Executive Director, 2007-2008, U.S. Environmental Protection Agency,
       Washington, DC 20460
Resha Putzrath, former Health Science Coordinator, U.S. Environmental Protection Agency,
       Office of the Science Advisor, Washington, DC 20460
ACKNOWLEDGMENTS
The technical assistance of Ms. Bette Zwayer is gratefully acknowledged.
          This document is a draft for review purposes only and does not constitute Agency policy.
5/11/11                                     ix       DRAFT: DO NOT CITE OR QUOTE

-------
 1                                     1.  INTRODUCTION

 2    1.1.  BACKGROUND
 3          Risk assessment is the U.S. Environmental Protection Agency (EPA or Agency)'s key
 4    process for deriving scientific information for making sound decisions on managing risks to
 5    human health and the environment (U.S. EPA, 2004). No risk assessment can reflect true risk
 6    with absolute certainty, so it is important that uncertainties be handled in a predictable,
 7    scientifically defensible way that is both consistent with the EPA's mission and responsive to the
 8    needs of decision makers (U.S. EPA, 2004). This involves decreasing uncertainty in estimates
 9    wherever possible, defining uncertainty and variability in estimates, and quantifying the
10    uncertainty when feasible. Historically, the Agency has used default uncertainty factors to
11    compensate for a lack of information. As science has advanced, however, there has been a
12    growing effort to increase reliance on the available data to modify the values for these
13    uncertainty factors (IPCS, 2005); this guidance describes an approach for using such information
14    for developing Data-Derived Extrapolation Factors (DDEFs).
15          An Examination of EPA Risk Assessment Principles and Practices: a Staff Paper
16    (U.S. EPA, 2004) notes that the EPA has published a number of documents that provide
17    direction in describing uncertainty in risk estimates including the following:
18
19
20       •  Risk Characterization Handbook (U. S. EPA, 2000)
21       •  Guiding Principles for Monte Carlo Analysis (U.S. EPA,  1997)
22       •  May 1997 Policy for Probabilistic Analysis (U.S. EPA, 1997a, b)
23       •  Risk Assessment Guidance for Super fund: Volume III—Part A, Process for Conducting
24          Probabilistic Risk Assessment (U.S. EPA, 2001 a)
25
26
27          Given that risk assessors never have a complete data set, it is accepted practice to use
28    default values and processes in order to allow a risk assessment to proceed in the absence of data.
29    The EPA uses the definition of default assumption articulated by the National Research Council
30    (NRC): "the option chosen on the basis  of risk assessment policy that appears to be the best
31    choice in the absence of data to the contrary" (NRC,  1983). The NRC, in its report Science and
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                               Page 1 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
 1   Judgment in Risk Assessment (NRC, 1994), supported EPA's use of defaults as a reasonable way
 2   to deal with uncertainty.  That report stated that the EPA should have principles for choosing
 3   default options and for judging when and how to depart from them.
 4          Since then, the EPA now initiates the process of choosing a method for developing
 5   uncertainty factors by evaluating the available data—guidance suggests invoking default values
 6   only when data are unavailable or insufficient. This contrasts with the previous position of using
 7   the strength of the data as the basis for moving away from default values for uncertainty factors.
 8   Specifically, the 2005 Guidelines for Carcinogen Risk Assessment (or "'Cancer Guidelines")
 9   (U.S. EPA, 2005) state "these cancer guidelines view a critical analysis of all of the available
10   information ...as the starting point from which a default option may be invoked if needed to
11   address uncertainty or the absence of critical information. " Thus, while risk assessors have
12   generally tried to make maximum use of available  data, the shift away from standard default
13   assumptions was codified as EPA science policy with the publication of the 2005 Cancer
14   Guidelines. Evaluating the available data will improve the scientific basis of risk assessments
15   when data are sufficient for refining uncertainty factors (UFs). In cases where data are not
16   sufficient,  hazard and risk characterizations will be improved, and data needs can be noted and
17   potentially filled in the future (Murray and Andersen, 2001; Meek, 2001; Meek et al., 2001;
18   Bogdanffy et al., 2001).
19
20   1.2.  PURPOSE AND SCOPE
21          U.S. and international efforts have improved the scientific basis for human health risk
22   assessments by increasing the use of mechanistic and kinetic data. For example, the EPA's 2005
23   Cancer Guidelines (U.S. EPA, 2005) emphasize the use of mode-of-action (MOA) information
24   in characterizing potential health effects of exposure to environmental agents.  International
25   efforts, including those by the International Life  Science Institute (ILSI) and the World Health
26   Organization (WHO)'s International Programme on Chemical Safety (IPCS), have developed
27   frameworks for evaluating animal data to determine the human relevance of described MO As
28   (Boobis et al., 2008;  Seed et al., 2005;  Sonnich-Mullin et al., 2001).  These documents guide the
29   qualitative and quantitative evaluation  of the relevance of a particular animal model of action in
30   humans and discuss the use of in vivo and in vitro  data when considering animal-to-human

                This document is a draft for review purposes only and does not constitute Agency policy.
     5/11/11                                Page 2 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
 1    extrapolation. The 2005 Cancer Guidelines (U.S. EPA, 2005), and other documents like IPCS's
 2    chemical-specific adjustment factors (CSAFs) guidance (IPCS, 2005), the Methods for
 3    Derivation of Inhalation Reference Concentrations and Application of Inhalation Dosimetry
 4    (U.S. EPA, 1994), and An Examination of EPA Risk Assessment Principles and Practices: Staff
 5    Paper Prepared for the U.S. EPA by Members of the Risk Assessment Task Force (U.S. EPA,
 6    2004) also encourage the use of sophisticated models like physiologically based pharmacokinetic
 7    (PBPK) and biologically based dose-response (BBDR) models in interspecies extrapolation.
 8           This document deals specifically with the development and use of data-derived factors in
 9    the calculation of nonlinear low-dose  estimates, or safety assessments.  The goal of DDEFs is to
10    maximize the use of available data and improve the scientific support for a risk assessment. The
11    processes described herein have benefited from the continuing discussion in the scientific
12    community on ways to replace the 10-fold uncertainty factors (10* UFs) that have historically
13    been used in deriving safety assessments such as reference doses (RfDs), minimal risk levels,
14    and acceptable daily intakes. WHO's IPCS guidance for deriving CSAFs was finalized in 2005.
15    This CSAF guidance describes approaches for use of kinetic and mechanistic data to refine inter-
16    and intraspecies extrapolation factors.  The IPCS guidance is largely based on analyses by
17    Renwick (1993) and Renwick and Lazarus (1998), which describe the use of toxicokinetic and
18    toxicodynamic data as a means of replacing the traditional  10* UFs for human sensitivity and
19    experimental animal-to-human extrapolation. This data-derived approach assigns values for
20    toxicokinetic and toxicodynamic differences as replacements for each traditional 10* UF.
21    Important distinctions between IPCS (2005) and the present EPA guidance are that IPCS restricts
22    toxicokinetic evaluations to the central compartment, disallowing local tissue metabolism to be
23    quantified as part of the toxicokinetic processes;  division of the animal  to human extrapolation
24    unevenly, attributing a greater fraction of default uncertainty to TK than to TD; and a general
25    level of depth.
26           The current document describes the EPA's approach to calculating extrapolation values
27    based on data; these are called data-derived extrapolation factors (DDEFs). DDEFs are  similar
28    in concept to IPCS/WHO's CSAFs in that the standard extrapolation factors are separated into
29    toxicokinetic (TK) and toxicodynamic (TD)  components, and kinetic and mechanistic data are
30    used to derive refined interspecies or intraspecies extrapolation factor(s). Conceptually, DDEFs
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                                Page 3 of 49    DRAFT: DO NOT CITE OR  QUOTE

-------
 1    (and CSAFs) may not be limited to a specific chemical but may also apply to chemicals with
 2    common structural characteristics, common MO A, or common toxicokinetic characteristics or
 3    determinants.  An appendix to this document contains  case study examples taken from the
 4    Integrated Risk Information System (IRIS) and from program office records.  These case studies
 5    present the application of principles contained in this document to data and modeling studies for
 6    actual chemicals and should serve as instructional aides.
 7           Topics most relevant to the derivation and use of DDEFs are the focus of this document.
 8    Thus, there are concepts beyond the scope of this guidance that are not discussed in detail here:
 9    approaches for selecting critical effects; establishing key events in an MOA analysis;1 deriving
10    points of departure; performing benchmark dose analysis; and developing and evaluating PBPK
11    and BBDR models. In addition, this document deals only with DDEF for the areas of inter-  and
12    intraspecies extrapolation; there is no discussion of factors that have been used for other areas of
13    uncertainty or variability (e.g.,  duration, database deficiencies, or lack of a
                                                 9
14    no-observed-adverse-effect-level [NOAEL]).
15
      1 Mode of action (MOA) refers to a series of key, determinant, and necessary interactions between the toxicant and
      its molecular target(s) that lead to the toxic response.  Refer to Section 2.2.4.1 for further information.
      2 Note:The Food Quality Protection Act (FQPA) mandates the use of a presumptive 10-fold factor for the protection
      of infants and children in addition to inter- and intraspecies factors.  This factor can only be modified based upon
      reliable data. The FQPA factor is not discussed in this document.
                 This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                                 Page 4 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
 1                 2.  TECHNICAL CONCEPTS AND PRINCIPLES FOR DDEF

 2    2.1.  BACKGROUND
 3          The methodologies of the EPA derivation of reference concentrations (RfCs) and RfDs
 4    (the predominant EPA nonlinear approaches) recognize steps for inter- and intraspecies
 5    extrapolation, both of which may include the application of uncertainty factors to an
 6    experimental result to account for recognized uncertainties in, and variability inherent in, the
 7    extrapolations from the experimental data conditions to estimates appropriate to the assumed
 8    human scenario (U.S. EPA, 2011, 2002b, 1994, 1993). This document describes an approach to
 9    performing inter- and intraspecies extrapolations based on the use of the best available science
10    and data.  DDEFs are factors estimated from quantitative data on interspecies differences or
11    human variability (illustrated in Figure 1).  DDEFs may consider both toxicokinetic and
12    toxicodynamic properties. These factors can be derived for a single agent or chemical, for a
13    class of chemicals with shared chemical or toxicological properties, and for a group of chemicals
14    that share a mode or mechanism of action or toxicokinetic characteristics. As described below,
15    DDEFs can be calculated using sophisticated toxicodynamic or toxicokinetic models or can be
16    calculated as ratios using key kinetic or dynamic data.  With regard to interspecies extrapolation,
17    the EPA currently recognizes a hierarchy of approaches ranging from the preferred approach
18    using PBPK modeling (U.S. EPA, 2006, 1994) down to default approaches for situations for
19    which data do not support an alternate approach, with DDEFs falling intermediate in this
20    hierarchy.
21          The default approach for the inhalation exposure route involves a combination of
22    application of a categorical dosimetric adjustment factor and a residual uncertainty factor
23    (U.S. EPA, 1994). The dosimetric adjustments are based on the following:
24
25       •  Anatomic and physiologic differences between species
26       •  Physical differences between particles and gases
27       •  Whether the toxic effect(s) are portal of entry or systemic in nature
28
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                                Page 5 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
  i
Il
OQ O
x a

o
 : TO ^
  >j

 i—> OJ
             cu
             V*
             a
             o
             &
             6ft
                 100 1
                  80-
                  60-
            U
                  40-
                  20-
                                                        /
                                                      /
                                                                                       Human

                                                                                       Animal
                                                                          Animal NOAEL
/
ilKf
'""I
e£—
UFA
'7
1
1
1 .^^"^ orBMD
_^. ^ jt*^
^ n^
.....^
10 100

	 1
1000
                                                          Dose
             Figure I.  Derivation of RfDs/RfCs using uncertainty factors.

-------
 1          For the oral exposure route, the default approach for interspecies extrapolation involves
 2    scaling the applied dosing, according to body weight, to the 3/4 power, and a residual uncertainty
 3    factor (U.S. EPA, 2011). Apportioning the default values for both inter- and intraspecies
 4    extrapolation is based on data for various chemicals. It is generally recognized that toxicokinetic
 5    data are more widely available than toxicodynamic data.  The magnitude of variation in the
 6    available TK data  suggests that the interspecies uncertainty factor might be evenly divided
 7    between TK and TD components. These values are /^ order of magnitude in value and can be
 8    seen in various documents as values of 3, 3.0, 3.16,  or 3.2.  Regardless of their values, the
 9    mathematical combination of two factors of !/2 order of magnitude each results in a value of
10    10 (i.e., 3x3 = 10). After quantifying TK differences between species, the residual uncertainty
11    factor associated with either route (oral or inhalation) has a default value of 3,  which may be
12    modified based on available data (U.S. EPA, 2011,  1994).  In accordance with the hierarchy of
13    approaches, when  available agent-specific data are supportive of DDEF derivation, a
14    data-derived approach is preferred over using the RfC approach or % body-weight scaling.
15
16    2.2.  EXTRAPOLATION WITH DDEFs
17          The foundation of DDEFs is the concept that the toxicity of a particular agent is due to a
18    combination of both toxicokinetic and toxicodynamic factors and that those factors can be
19    quantified in animals and humans. For purposes of this guidance, toxicokinetics (TK) is defined
20    as the determination and quantification of the time course and dose dependency of absorption,
21    distribution, metabolism, and excretion of chemicals (sometimes referred to as pharmacokinetics
22    or ADME). Toxicodynamics (TD) is defined as the determination and quantification of the
23    sequence of events at the cellular and molecular levels leading to a toxic response.  There is no
24    clear separation between TK and TD because the processes leading to biological responses
25    include aspects of both—including interactions between TK and TD processes.
26
27    2.2.1. Approaches to Deriving DDEFs
28          The focus of this guidance is on extrapolation from  animals to humans, and within the
29    human population. Extrapolation can be accomplished by one of several approaches ranging
30    from the use of highly sophisticated BBDR models to the calculation of relatively simple ratios
31    using TK or TD data describing critical factors in inter- or intraspecies extrapolation.  The
                 This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                                Page 7 of 49    DRAFT:  DO NOT CITE OR QUOTE

-------
 1    following text describes these approaches.  In the absence of data for performing sophisticated
 2    modeling or for deriving DDEF values, default approaches are used. Figure 2 is a flowchart
 3    depicting the decision process used in deriving and applying extrapolation factors.
 4
 5    2.2.1.1.  TK and TD Models
 6          TK and TD models represent the preferred approach to intra- and/or interspecies
 7    extrapolation. They vary in level of complexity from classical compartmental and simple
 8    statistical response models to physiologically realistic models of TK and TD processes, up to and
 9    including BBDR models.  These models provide a quantitative description of the biological
10    processes involved in the toxicokinetics and/or MO A of chemical(s).  In these TK and TD
11    models, some measure of the internal dose is related to the external dose and mode of action,
12    respectively.
13          TK modeling is the process of developing a mathematical description of ADME in a
14    living organism. Two common types of models are (1) data-based classical noncompartmental
15    or compartmental models and (2) PBPK models. Data-based models, also known as classical
16    models, mathematically describe the temporal change in chemical concentration in blood, tissue,
17    or excreta of the species in which the data were generated. The classical models treat the body
18    as a single homogenous or multicompartment system with elimination occurring in a specific
19    compartment; the characteristics of the compartments (number, volume, etc.) are hypothetical in
20    that they are chosen for the purpose of describing the data rather than a priori based on the
21    physiological characteristics of the organism.  Due to these characteristics, classical models are
22    used for interpolation, i.e., within the range of doses,  dose route, and species in which the data
23    were generated (Renwick, 1994).
24          PBPK models differ from classical compartmental models in that they are composed of
25    compartments with realistic tissue volumes that are linked by blood flow. Other parameters used
26    in these models account for chemical-specific characteristics that can be independently measured
27    in both humans and laboratory animals (usually using in vitro techniques); these
28    chemical-specific parameters include tissue solubility (i.e., partition coefficients), binding, and
29    metabolism.  These models are used to simulate the relationship between applied dose and
30    internal dose.  They are more data intensive to develop compared to classical compartmental

                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                                Page 8 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
     Data-Derived
     Extrapolation
     Factors
                                        Review of
                                        Available
                                         Data
                                            Do the data inform
                                            inter-ortntraspecies
                            Animal to
                             Human
                           Extrapolation
                                                          witnin Human
                                                           Variation
                                                           Use
                                                          model
                                                          forEF,,,
                                                          and/or
                                                           EF
                                Use
                               model
                               for EFAK
                               and/or
                                EFAD
                                                                             Yes, only TK
                                                                             or only TD
          Yes, only TK
          or only TD
                   Use data
                   forEF
                    OR
                    EF
                                                                       Use data
                                                                       forEF
                                                                        OR
                                                                        EF
                                                                                         efau
                                                                                       procedure
                                                                                         for
                                                                                       remaining
                                                                                       ncertaint
 Default
procedure
   for
remaining
uncertainty
Composite Factor (CF)
2
O

4

5

6

7
    Figure 2. Decision process for DDEFs. The availability of an adequate
    pharmacokinetic (PK) or pharmacodynamic (PD) model is first considered followed by
    analysis of the availability of adequate data to describe the toxicokinetics (TK) and/or the
    toxicodynamics (TD) of the chemical. With the availability of an adequate model or
    data, data-derived extrapolation factors for intraspecies (£FAK, EFAD) and interspecies
    extrapolation (EFnK, EFno) are developed.  Such data-derived  factors are preferred over
    default factors.  In the absence of an adequate model or data, default factors are used.


    *For interspecies extrapolation,  the default procedure is % body-weight scaling for oral
    (U.S. EPA, 2006) and the RfC method (U.S. EPA,  1994) for inhalation to account for
    potential TK differences with a 3 x factor for potential TD differences.  The composite
    factor (CF) accounts for inter- and intraspecies extrapolation and can comprise default or
    DDEF values for the four extrapolation factor components.
     5/11/11
        This document is a draft for review purposes only and does not constitute Agency policy.

                                     Page 9 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
 1    models, but they are advantageous because they can be used for extrapolation (i.e., across dose
 2    range, among animal species, between routes of exposure, and across exposure scenarios)
 3    (Krishnan and Andersen, 1994; U.S. EPA, 2006).
 4           TD models can be developed when there are sufficient data to both ascertain the MOA
 5    and to quantitatively support model parameters that represent rates and other quantities
 6    associated with key precursor events in the MOA.  A BBDR model describes biological
 7    processes at the cellular and molecular levels in such a way as to link target tissue dose with
 8    adverse effect; in practice, BBDR models are often described as a combined TK/TD model.
 9    These models may be used for extrapolation. However, with adequate understanding of the
10    nature of the response, empirical data describing the dose-response function in relevant species
11    or population groups are sufficient to serve as the basis for DDEF derivation; in these cases, a
12    fully developed TD model may not be required.
13           DDEF values are extrapolation factors, as opposed to uncertainty factors, per se. DDEF
14    values are quantitatively derived based  on TK and/or TD data for the chemical under evaluation.
15    DDEF values are not the same as the default uncertainty factor values, but the values for the
16    DDEF components may sometimes be similar to default values for uncertainty factors.
17    Developing a DDEF value reduces uncertainty and carries with it a change in nomenclature.
18
19    2.2.1.2. Use of Ratios to Calculate DDEF
20           In the absence of sufficient data to develop a robust TK or TD model, the risk assessor
21    need not necessarily use default 10x UFs. DDEFs can be calculated as ratios using data from
22    key studies evaluating TK or TD profiles or properties of a particular chemical. Example
23    equations for calculating DDEFs are provided in Table 1 and are described in more detail in
24    Sections 3 (TK) and 4 (TD).
25           In general, interspecies extrapolation involves calculating a ratio of human data for a
26    kinetic or dynamic parameter to animal data for a kinetic or dynamic parameter. Similarly, for
27    intraspecies extrapolation, a ratio is calculated using data from the sensitive population and that
28    for the general, or average, population.  Data to derive the TK factors may come from in vivo or
29    in vitro studies.  For TD, in general, interspecies extrapolation may come  from in vivo studies
30    but will often be accomplished with in vitro data in a relevant tissue.  When data on toxic effects

                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                               Page  10 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
             Table 1.  Example equations used to derive DDEFs
Extrapolation
Animal to human
(interspecies)
Within human
(intraspecies)
Toxicokinetics
(see Section 3)
Dose,
FF
1^1 AT,"
**- DoseH
AUC
pp _ gen
•'-'-THE-
Aiir
AUL-%tile
Toxicodynamics
(see Section 4)
Concentration,
EF -
^ ConcentrationH
Concentration pn
EF
110 Concentrationsens
 4
 5
 6
 7
 8
 9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
DoseA
DoseH
EFAD
ConcentrationA
ConcentrationH
EFnK
AUCgen
AUCsens
EFm
Concentration^,!
: extrapolation factor for interspecies extrapolation covering toxicokinetics.
: administered or external dose to the animal.
: administered or external dose to the human.
: extrapolation factor for interspecies extrapolation covering toxicodynamics.
: concentration of the agent at the tissue in the animal.
: concentration of the agent at the tissue in the human.
: extrapolation factor for intraspecies extrapolation covering toxicokinetics.
: area under the curve at a measure of central tendency in the general human population.
: area under the curve at a percentile of interest in the human population.
: extrapolation factor for intraspecies extrapolation covering toxicodynamics.
: concentration at a measure of central tendency in the general human population.
: concentration at a percentile of interest in the human population.
are available in humans, these data may be used directly for the point of departure (POD)
development, eliminating the need for the interspecies extrapolation. Likewise, they can be used
to inform an interspecies factor when the POD is derived from animals.
       For DDEFs involving interspecies extrapolation, it is preferred that the ratio be based on
data at or near the POD.  When sufficient data are available, DDEF values should be calculated
for a range of doses near the POD because the shape of the dose-response curve can vary among
species.  Metabolism and kinetic properties can vary across doses, particularly in  the higher dose
ranges; thus, using estimates at or near the POD helps avoid introducing significant uncertainty
in the DDEF estimate caused by nonlinearity in kinetic properties.  Evaluating a range of PODs
takes into account the variability of the DDEFs based on the POD selected.  The interspecies
DDEF should be derived using an estimate of central tendency, such as the mean, median, or
mode,  depending on the characteristics of the  data. It is, however, important to evaluate
      5/11/11
           This document is a draft for review purposes only and does not constitute Agency policy.
                                        Page 11 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
 1    variability in the DDEF. Thus, it is recommended that, to the extent possible, the hazard and risk
 2    characterizations reflect the upper and lower confidence bounds on the DDEF.
 3          By contrast with interspecies extrapolation, when calculating intraspecies DDEFs, the
 4    ratio involves consideration of a measure of central tendency of the general population and lower
 5    percentiles of interest (e.g., 1st, 2.5*, or 5*) to represent the sensitive populations. As the needs
 6    of risk managers and decision makers vary, it is recommended that a range of percentiles be
 7    evaluated and reported in the hazard and risk characterizations.
 8          Toxicokinetic ratios (for either interspecies or intraspecies extrapolation) are based upon
 9    the relevant dose metric, such as area under the curve (AUC) and the maximum concentration
10    (Cmax).3  Other metrics (e.g., AUC  above  a threshold) may be used if supported by the data or if
11    relevant for a particular chemical or MOA.  For toxicants that bind covalently or cause
12    irreversible damage, especially as a consequence of subchronic or chronic exposure, an
13    integrated measure of dose over time such as AUC is generally used (O'Flaherty, 1989). In the
14    case of effects occurring as a consequence of acute exposure, Cmax may be more appropriate
15    (Boyes et al., 2005; Barton, 2005).  When data on chemical-specific AUC, Cmax,  or clearance
16    (Cl) are not available, a chemical-related  physiological parameter (e.g., renal glomerular
17    filtration rate) that is critical to the onset of toxicity or to the MOA may be used.
18          As Table 1 indicates, there  are generally four DDEFs that can be calculated, given
19    sufficient information.  Two are for extrapolation from animal data to humans: EFAK is
20    calculated to account for TK variability, while  EFAo deals with TD variability. Likewise, there
21    are two factors dealing with variability within the human population: EFnK for TK and EFno for
22    TD. Table 1 provides example equations for calculating these DDEFs. Section 3 describes
23    specifics for TK factors for interspecies (see Section 3.2) and intraspecies (see Section 3.3)
24    extrapolation.  Section 4 describes TD factors for both animal to human (see Section 4.2) and
25    within human (see Section 4.3) extrapolation.  Section 5 describes how to combine the EFAK,
26    £FAD, EFnK, and EFno into the composite UF.
27          The overall goal of DDEFs is to maximize the use of available data and improve the
28    overall scientific support for a risk assessment. Figure 2 provides a flowchart of the decision
29    process for extrapolation used in deriving DDEFs.  As shown in the figure, inter- and
      3 Clearance can be used to calculate this ratio when it can be assumed or demonstrated that the relevant dose metric
      is AUC or concentration at steady state.
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                                Page 12 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
 1    intraspecies extrapolation can be accomplished using a combination of TK or TD models,
 2    DDEFs derived from ratios, and/or use of defaults.  As described in more detail in Sections 3 and
 3    4, it is important for the hazard and/or risk characterizations to include thorough and transparent
 4    discussions of methods and data used to support extrapolation approaches.
 5
 6    2.2.2. Qualitative Considerations
 7          Although in some cases there may not be sufficient data for a quantitative estimate of a
 8    DDEF, there may still be information to support a UF different from the default. For example,
 9    there may be qualitative evidence that a MOA identified in animals is not relevant to humans. A
10    framework developed by ILSI for evaluating the relevance of an animal MOA can be found in
11    Seed et al. (2005), Meek et al.  (2003), and Boobis et al. (2008).  The human relevance
12    framework provides a transparent and logical thought process by which animal and human MOA
13    data can be  evaluated on both a qualitative and quantitative basis.  In these cases, where only
14    qualitative data are available, a thorough weight-of-evidence analysis should be considered with
15    the hazard and/or risk characterization to discuss the derivation of the DDEF along with
16    associated uncertainties in the  available database.
17
18    2.2.3. Information Quality
19          Critical evaluation of all data used to support the development of DDEFs is necessary.
20    This includes data used to provide qualitative support for the MOA and choice of dose metric, as
21    well as data used in the quantitative derivation of the DDEF itself.  Supporting studies can be
22    evaluated using criteria set forth in various EPA guidance documents, including the recently
23    published 2005 Cancer Guidelines, as well as earlier guidelines specific to neurotoxic,
24    reproductive,  and developmental endpoints (U.S. EPA, 1998, 1996, 1991). In addition, the
25    general principles outlined in the EPA information quality guidelines are applicable in the
26    critical evaluation of data used to support DDEF development (U.S. EPA, 2002a). The
27    remainder of this section highlights some areas of special emphasis that are particularly relevant
28    to the DDEF derivation process including MOA, uncertainty and variability, and dealing with
29    multiple responding organs or tissues.
30          Use of secondary data sources is one particular area of concern. Examples of secondary
31    data sources include compilations of pharmacokinetic parameters (e.g., Brown et al., 1997) and
                 This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                               Page 13 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
 1    studies cited and summarized in toxicity profiles and review articles.  In general, for principal
 2    and supporting studies used directly in DDEF derivation, review of the original literature is
 3    necessary. In the case of critical assumptions and data, contradictory results from different
 4    studies are best resolved by review of the original publications.
 5           Quantitative TK and TD data used in the DDEF-derivation process requires particular
 6    attention to the appropriateness of the study design, the analytical methodology used, and the
 7    statistical analysis of the data. Consideration of appropriate study design extends beyond simply
 8    verifying that the methods used were adequate for the goals of the study; it also encompasses
 9    consideration of the relevancy of the animal species or in vitro test system to evaluate MO A.
10    Relevance can be assessed in both qualitative and quantitative terms.  For example, if there is a
11    lack of species concordance (i.e., a particular TK or TD process does not occur in humans) or
12    effects occur only under physiologically unrealistic conditions or not in the tissue evaluated, then
13    its relevancy is questionable and uncertain. Criteria used in arriving at such a determination have
14    been published for both the more general case (Seed et al., 2005) and particular endpoints
15    including various forms of rodent cancer (Proctor et al., 2007; Maronpot et al., 2004). Particular
16    considerations relevant to the use of in vitro data are discussed below. Another important factor
17    in terms of relevancy is consideration of whether the TK or TD response represents a uniquely
18    susceptible tissue, process, or population.  This is a critical determinant in evaluating the use of
19    data to describe intraspecies variability.
20
21    2.2.4. Additional Considerations
22    2.2.4.1.  Mode of Action
23          Information on MOA can greatly enhance DDEF derivation, even when a complete
24    explication of mechanism is not available.  In the 2005 Cancer Guidelines, the EPA describes
25    MOA evaluation as the critical information that defines the conditions under which a toxicant
26    causes its effect, the relevance of animal data for hazard identification, and the most appropriate
27    approach to low-dose extrapolation.  The 2005 Cancer Guidelines also present a framework for
28    evaluating data in support of MOA determination.  Major components of this framework include
29    a description of the hypothesized MOA and a discussion of the experimental support for the
30    hypothesized MOA based on modified Hill criteria (U.S. EPA, 2005) for demonstrating
31    associations  in human studies.
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                                Page 14 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
 1           MOA is defined as a sequence of key events and processes, starting with the interaction
 2    of an agent with a cell, proceeding through operational and anatomical changes, and resulting in
 3    toxicity. A key event is an empirically observable precursor step that is itself a necessary
 4    element of the MOA or is a biologically based marker for such an element. MOA is contrasted
 5    with "mechanism of action," which implies a more detailed understanding and description of
 6    events, often at the molecular level, than is meant by MOA (U.S. EPA, 2005).
 7           DDEFs for both TK and TD are endpoint driven—that is, considered in the context of the
 8    toxic endpoints most relevant for purposes of the risk assessment. Understanding MOA for the
 9    agent(s) of interest helps to ensure that the TK or TD parameter used to derive the DDEF will be
10    robust scientifically.  The key events in MOA are likely to identify important metabolite(s) and
11    potential species differences. Moreover, data on key events may be used directly to estimate the
12    EFAK or the EFAo-
13
14    2.2.4.2. Use of In Vitro Data
15           In vitro assays play an important role in defining DDEFs; however, care must be taken to
16    avoid taking isolated findings out of context. Consideration of interspecies differences in ADME
17    is essential because the dose to target tissue in any given exposure scenario is a balance among
18    multiple and competing ADME processes. Thus, in vitro data should not be used for quantitative
19    purposes unless interpreted in the context of the intact system. Among the questions to be
20    considered when applying in vitro data to DDEFs are the following:
21
22
23       •   Was the lexicologically active form of the agent studied?
24       •   How directly was the measured response linked to the toxic effect?
25       •   Are the biological samples used in the assays derived from equivalent organs, tissues, cell
26           types,  age, stage of development, and sex of the animals/humans in which the target
27           organ  toxicity was identified?
28       •   What is the range of variability (e.g., diverse human populations  and lifestages) that the
29           biological materials cover?4
      4 Quality (purity, viability, source) of the samples is of particular concern, with biological materials derived from
      human organ donors.
                 This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                                Page 15 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
 1
 2
 3
 4
 5
 6
 7
 9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
    •  If the effect occurs or can be measured in several tissues, is the studied tissue or tissue
       preparation an appropriate surrogate?  Or, in situations where the effect is not localized,
       is the effect consistent across tissues?
    •  Does the design of the study allow for statistically valid comparisons based on such
       factors as replication and sample size?
    •  Was chemical uptake considered when the chemical was applied to the samples so as to
       give comparable intracellular concentrations across tissues, and similar tissues across
       species?
    •  Do the concentrations in the in vitro studies allow for comparison with in vivo
       conditions?
All of these issues affect the utility of applying in vitro data for risk assessment: a clear
discussion of these points helps to clarify the appropriateness of the information used for
deriving DDEFs.
2.2.4.3.  Uncertainty and Variability
       The application of the inter- and
intraspecies UFs attempts to account for both
the variability (true heterogeneity) and
uncertainty (lack of knowledge) the in the data
available (see Textbox 1, U.S. EPA, 2002b).
The DDEFs described in this document
evaluate variability within the data. Evaluation
of the sources and magnitude of uncertainty is
appropriate (U.S. EPA, 2005, 2001, 1997a,  b).
Quantitative uncertainty analyses may be
                   Textbox 1
Variability refers to true heterogeneity or diversity.
This may be due to differences in exposure as well as
differences in response. Those inherent differences are
referred to as variability. Differences among
individuals in a population are referred to as
interindividual variability, while differences for
one individual over time are referred to as
intraindividual variability.
Uncertainty occurs because of lack of knowledge. It is
not the same as variability. Uncertainty can often be
reduced by collecting more and better data, while
variability is an inherent property of the population
being evaluated.  Variability can be better characterized
with more data but cannot be eliminated. Efforts to
clearly distinguish between variability and uncertainty
are important for both risk assessment and risk
characterization.
Source: U.S. EPA (2002b).
undertaken but are not presented in this document. When quantitative approaches are not
feasible, qualitative uncertainty analyses may be developed. As is consistent with the 2005
Cancer Guidelines: "a default option may be invoked if needed to address uncertainty or the
absence of critical information. "
      5/11/11
            This document is a draft for review purposes only and does not constitute Agency policy.
                                         Page 16 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
 1    2.2.4.4.  Multiple Critical Effects
 2          For some toxicants, multiple critical effects may be identified during hazard
 3    identification. In some cases, these effects may be the result of a single MOA. However, for
 4    others, the critical effects may have different or unknown MO As.  It is possible that the
 5    uncertainty and/or variability associated with the TK and/or TD of each effect may differ,
 6    resulting in different DDEFs.  The results generated for the multiple responding tissues/organs,
 7    particularly if multiple MOAs are operational or MOA is unknown, should be presented for
 8    comparison (for example, in a table that is accompanied by a discussion of the methods used).
 9    Unless there is scientific support for doing so, it is important not to mix DDEFs derived for one
10    tissue or one MOA with DDEFs derived from a different tissue. For example, DDEF values for
11    kidney effects may not apply to liver effects.
12
13    2.2.4.5.  Screening-Level vs. Refined Risk Assessments
14          Extrapolation is most scientifically robust when data are first evaluated prior to the use of
15    defaults. However, with a multitude of types of data, analyses, and risk assessments, as well as
16    the diversity of needs of decision makers, it is neither possible nor desirable to specify
17    step-by-step criteria for decisions to invoke a default option.  Some risk assessments may be
18    limited by time or resource constraints. Other risk assessments may provide only screening level
19    evaluations. In these cases, the risk assessment may be more likely to resort to one or more
20    default assumptions. On the other hand, risk assessments used to  support significant risk
21    management decisions will often benefit from a more comprehensive assessment.
22
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                                Page 17 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
 1                       3.  DDEFs BASED ON TOXICOKINETICS (TK)

 2    3.1.  GENERAL CONSIDERATIONS
 3          Toxicokinetics is concerned with delivery of the biologically active chemical species to
 4    the target tissue of interest. Data on tissue concentrations of toxicants or clearance rates of
 5    toxicant removal serve as the basis for deriving extrapolation factors foor toxicokinetic
 6    components. This section provides a discussion of factors common to derivation of both inter-
 7    and intraspecies uncertainty factors to account for TK variability. Data on the quantitative
 8    differences in the TK between animals and humans  are used for interspecies extrapolation
 9    (£FAK); differences in susceptibility within the human population are used for the intraspecies
10    extrapolation (£FHK)- Thus, the factor £FAK accounts for extrapolation from laboratory animals
11    to the general human population. The £FHK factor accounts for the variation in the
12    dose/exposure-response relationship between the general human and potentially susceptible
13    human individuals or groups. Note, the term susceptible is also used to describe sensitive
14    individuals or groups, as these two terms are often used interchangeably, and no convention for
15    their use is widely accepted (U.S. EPA, 2004).  Developing a DDEF for TK requires knowledge
16    about the relationship between external dose and internal (target tissue) concentrations. This
17    information can come from studies in which tissue concentrations are observed, both types of
18    data are recorded, or can come from adequate TK models, which expand the range of confidence
19    from  that of the empirical observations. TK models, especially PBPK models, represent an
20    important tool through which in vitro observations can be interpreted in the context of the intact
21    system. As such, they represent an advantageous means to evaluate the impact of studies
22    (especially those using human tissues) conducted in vitro.
23          The TK portion of each factor (£FAK, EFRK) is combined with the corresponding TD
24    factors to assemble the composite extrapolation factor (see Section 5). Where the data are not
25    sufficient to derive  a DDEF for TK,  other approaches can be considered for £FAK or £FHK- For
26    example, the RfC approach (U.S. EPA, 1994) when evaluating inhalation data or % body-weight
27    scaling, or a default as described in Figure 2.
28          Important questions to address for TK are given below:
29
30       •   What is/are  the critical effect(s) and POD being used for this assessment?
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                               Page 18 of 49   DRAFT:  DO NOT CITE OR QUOTE

-------
 1       •  What is the MOA or mechanism for that toxicity? Have the key events been identified
 2          and quantified?  Do these key events identify important metabolic steps?
 3       •  Are the process of absorption, distribution, metabolism, and elimination of the chemical
 4          well characterized? Do animals and humans metabolize the chemical(s) in a similar way
 5          (qualitatively and quantitatively)?
 6       •  Are there data in human populations describing variation in important kinetic
 7          parameter(s) for this chemical(s)? Do these data identify a susceptible population(s) or
 8          lifestage(s)? Can the degree of this susceptibility be estimated?
 9
10
11          TK data may be developed empirically or through compartmental or physiologically
12    based TK models. Section 2.2.2 describes how data, models, and approaches are evaluated for
13    their appropriateness. For each critical effect identified for a particular agent, separate DDEF
14    analyses are conducted for EFAK and EFHK- As such, data for multiple susceptible
15    tissues/endpoints can be evaluated, concentrating on those tissues that demonstrate adverse
16    responses near the POD for the critical  effect.
17
18    3.1.1. Dose Metric
19          Dose metric is a term used to identify a measure of the internal dose that is associated
20    with the health outcome of interest. It describes target tissue exposure in terms of the toxic
21    chemical moiety (parent or metabolite) and is expressed in appropriate time-normalized terms.
22    For example, acute effects are often most related to peak concentrations, whereas  effects
23    occurring following a prolonged exposure are often best correlated with time-normalized (e.g.,
24    area under the concentration-time curve) measures of exposure.  The choice of the dose metric is
25    an important component in TK extrapolations. This choice depends on whether toxicity is best
26    ascribed to a momentary or transient tissue exposure or a cumulative dose to target tissue.  For a
27    given chemical, the appropriate dose metric will also be determined by,  and can vary with, both
28    the duration of exposure and the adverse effect of concern (U.S. EPA, 2006).  Selection of an
29    appropriate dose metric based upon specific endpoints involves several elements including those
30    described in more detail below:
31
32       •  Duration of exposure and effect;
33       •  Identification of the active chemical moiety;
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                                Page 19 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
 1       •   Selection of the organ or tissue group in which some measure of internal dose is desired;
 2           and
 3       •   Selection of the measure of exposure that best correlates with toxicity.
 4
 5
 6           Whether an adverse effect is a consequence of acute- or chronic-duration exposure
 7    impacts the choice of dose metric.  For acute, reversible effects (e.g., sensory irritation, narcosis),
 8    a measure of instantaneous or peak tissue exposure such as Cmax may be the most appropriate
 9    dose metric (Alarie, 1973; Boyes et al., 2005).  For chronic effects, in the absence of MOA
10    information to the contrary, it is generally assumed that some integrated cumulative measure of
11    tissue exposure to active toxicant is the most appropriate dose metric, e.g., area under the curve
12    (AUC). Alternative choices such as amount of chemical or rate of metabolite production can be
13    used as appropriate for a particular agent or MOA (U.S. EPA, 2006). For example, there may be
14    a case where a temporally large influx of active chemical to a target site in a relatively short
15    period of time (peak exposure)  is observed, in which case, a less commonly used metric such as
16    time above a critical concentration (TACC) may be most appropriate. In such an instance, the
17    data and rationale in support of a particular dose metric need to be presented.
18           Clearance, while not typically considered a dose metric, can be useful in DDEF
19    derivation. Clearance is mathematically inversely related to AUC (e.g., AUC = dose/clearance);
20    thus, differences  in clearance values can be used in calculation of ratios. When metabolism
21    represents the primary or sole clearance mechanism, either of two clearance models may be
22    applicable. Intrinsic clearance (Clint) has been  used for interspecies scaling of administered
23    doses in drug development (Houston and Carlile, 1997). Clint is calculated as Vmax/Km, and is in
24    units of volume cleared of the substrate per unit time. Vmax is the theoretical maximal initial
25    velocity of the reaction, and Km is  the substrate concentration driving the reaction rate at one-half
26    Vmax.  Clint can be extrapolated  to the whole body with knowledge of protein binding and the
27    recovery of the protein or cellular or subcellular fraction used in the in vitro investigations
28    (Carlile et al., 1997). Hepatic clearance (Clnep) is also based on Vmax/Km measurements but
29    includes a substrate delivery term,  whose value is governed by hepatic blood flow. These
30    measures of clearance differ in  that Cl;nt is not bounded by hepatic blood flow, but Clhep cannot
31    exceed hepatic blood flow. While metabolic rate constants (Vmax and Km) derived from in vitro
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                               Page 20 of 49   DRAFT:  DO NOT CITE OR QUOTE

-------
 1    data can also be scaled up and incorporated into PBPK models, the use of these clearance models
 2    is a simpler approach, useful when an appropriate PBPK model is unavailable.  Classical,
 3    compartmental TK analyses and measures of clearance are best suited for conditions where
 4    metabolism represents a detoxication process, when substrate concentration is less than the Km
 5    value, and when metabolism represents the major clearance mechanism.
 6          Whether toxicity is attributable to a parent chemical, a metabolite, or some combination
 7    of metabolites is a critical consideration.  The active chemical moiety can be identified through
 8    studies in which the toxicities induced by the parent chemical and metabolite(s) are compared or
 9    from the results of studies using enzyme inhibitors and/or inducers.  In vitro studies can also be
10    quite useful in this regard under appropriate conditions (see Sections 2.2.3.2 and 3.1.3).
11    Quantifying differences in dosimetry can be difficult when metabolic pathways become complex
12    (e.g., where competition among pathways may be concentration dependent). If the metabolic
13    pathway bifurcates and the identity of the bioactive metabolite(s) are unknown or unquantifiable,
14    determination of the appropriate dose metric can be highly uncertain.
15          The organ  or tissue group where the toxic effects occur is ideally the site from which
16    estimates of internal dose (tissue concentration) are generated. In practice, this information may
17    be unavailable in the absence of an appropriate PBPK model. It may be  necessary to use
18    absorbed dose of the parent chemical as a surrogate measure of internal dose. Another surrogate
19    dose metric is measurement of parent chemical or active metabolite in circulating blood if the
20    relationship between target tissue dose and blood is known or can be reliably inferred from
21    experimental data. Some data have demonstrated that blood:air partition-coefficient values may
22    vary appreciably between species but that tissue:air (e.g., liverair) partition coefficients are
23    similar among mammalian species (Thomas, 1975). It seems reasonable to use the cross-species
24    similarity in the primary determinant of diffusion from blood into tissues as a justification to rely
25    on concentrations  of the toxicant in blood as a surrogate for tissue concentrations.  However,
26    when local tissue bioactivation may be a determinant of the toxic response,  this should be given
27    careful consideration.  Those issues notwithstanding, measures of internal dose in circulating
28    blood (see IPCS, 2005) may be used as the basis for DDEF derivation under either of these
29    conditions:
30
31
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                               Page 21 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
 1       •   When evaluating interspecies differences, the distribution from blood to sensitive
 2           (critical) tissues is shown to be or can be assumed to be the same between animals and
 3           humans.
 4       •   When evaluating intraspecies differences, the distribution from blood to sensitive
 5           (critical) tissues is shown to be or can be assumed to be the same between members of
 6           the general human and potentially sensitive human groups.
 7
 8
 9           Because few data are available for concentrations of toxicants in human solid
10    tissues—such as liver, kidney, etc.—compared with data describing toxicant concentrations in
11    human blood, model predictions for solid tissue compartments are less certain than predictions of
12    toxicant concentrations in blood.  Partitioning of the active chemical from blood into systemic
13    target tissues may be governed more by physicochemical than by biological processes. This may
14    be considered another basis for relying on data describing the concentration and variability of the
15    biologically active metabolite in the central compartment.5 For example, the ratio of blood lipid
16    to tissue lipid concentrations may be a key determinant in the diffusion of lipophilic compounds
17    out of blood; however, differences in tissue lipid composition between species may be fairly
18    small compared to differences in blood flow and metabolic activity.
19
20    3.1.2.  Dose Selection
21           Because variability in internal dosimetry may be a function of dose, the selection of the
22    external exposure (inhaled concentration or orally ingested dose) is important. In cases where
23    toxicokinetics is nonlinear, the dose selected for the DDEF derivation will impact the magnitude
24    of £FAK or £FHK.  Using a dose at or near the POD alleviates some concerns regarding
25    nonlinearities in metabolism. Alternatively, data that show a linear relationship between external
26    dose and internal dose metrics can indicate generalizability of the £FAK or £FHK to doses that
27    may be higher or lower than those used in its calculation.
28
29    3.1.3.  In Vitro Data
30           In vitro techniques are important tools in evaluation of toxicokinetics as information can
31    be gathered that are impractical or unethical to collect in the intact animal or humans. However,
      5 The central compartment is defined as blood, plasma, or serum in the systemic circulation. All tissues except those
      representing the portal of entry are defined as peripheral compartments.
                 This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                                Page 22 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
 1    it is important when deriving EFAK and £FHK to consider interspecies differences in ADME. In
 2    vitro data should used for quantitative purposes only when interpreted in the context of the intact
 3    system, as discussed in Section 2.2.4.2.  Care must be taken to avoid taking isolated findings out
 4    of context.
 5
 6    3.2.  INTERSPECIES TOXICOKINETIC EXTRAPOLATION (EFAK)
 7         This section provides a discussion of the quantitative differences in the TK between
 8    animals and humans that are used for interspecies extrapolation (EFAK).  In this process,
 9    toxicokinetic differences between species are characterized as the ratio of applied doses in the
10    test species of interest and humans that result in the same level of the internal dose metric (see
11    Figure 3).  These values for the dose metric may be calculated from the external doses actually
12    used in the dose-response evaluations, or by normalizing the dose metric to account for
13    administered dose (e.g., correcting AUC for each species by dividing the AUC by the external
14    dose) when the relationship between the values for the dose metric value and the applied dose are
15    linear in the range of extrapolation.  Illustrative case studies are included in the appendix.
16
17    3.2.1. Considerations for EFAK
18          Extrapolation of TK data obtained in animal models to humans (EFAK)  is evaluated for
19    each potential critical effect of interest. Data for multiple susceptible tissues/endpoints are
20    evaluated, concentrating on those tissues that demonstrate adverse responses near the POD for
21    the critical effect.  This analysis includes consideration of MO A, identification of the active
22    chemical agent for this particular effect, and determination of the appropriate dose metric.  This
23    is evaluated as a weight-of-evidence approach emphasizing both qualitative and quantitative
24    evidence.  An important part of this process is evaluating concordance of metabolic processes
25    between the animal model and humans.  An additional consideration is whether the kinetic data
26    are from a "typical" or average adult animal as opposed to an animal model system that may be
27    unusually sensitive for a particular effect (e.g., metabolic knockout).
28
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                                Page 23 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
                                 ToxJcokinetics
              Applied Dose
                                              EF
                                                 AK
                                                               Applied Dose
2
3
4

5

6
                            Time
                                                                   Time
           Figure 3. Interspecies toxicokinetics.  In keeping with guidance set for inhaled
           substances (U.S. EPA, 1994), interspecies differences in toxicokinetics are defined as
           differences in the external dose producing the same level of the dose metric in the target
           tissue of interest in test animals and in humans representative of the general population.
           Because it is the default assumption of the Agency that humans are considered more
           sensitive than animals, EFAK is the ratio of animal dose:human dose suitable for
           developing a value consistent with its placement in the denominator (i.e., reference value
           = POD/UF).  For clarity in presentation, £FAK is applied to the external dose in animals.
       Furthermore, it is important to assess the relationship of externally applied dose to

internal dose metric over the entire range of dose levels used in the critical study.  There should

be careful attention paid both to measures of central tendency and to variability, particularly in
    5/11/11
          This document is a draft for review purposes only and does not constitute Agency policy.
                                     Page 24 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
 1   the range of concentrations or doses close to the point of inflection (where the shape or slope of
 2   the dose response curve changes) because of potential nonlinearities in metabolism.
 3
 4   3.2.2.  Computation
 5          For TK extrapolation, the goal is to determine differences in dosimetry between animals
 6   and humans.  For interspecies extrapolation, toxicokinetically equivalent exposures (doses) are
 7   determined by fixing the internal dose (level of the dose metric at or near the POD) and
 8   determining the ratio of external (applied) dose that results in the same level of the dose metric in
 9   animals and humans. This approach is consistent with that in the RfC guidance for inhalation
10   toxicants (U.S. EPA, 1994).
11
12   3.2.2.1.  Use of TK Models
13          A PBPK or other TK model provides the most biologically appropriate approach for
14   evaluating interspecies  TK extrapolation. The model is  subject to evaluation as previously
15   described (U.S. EPA, 2006). The model can be used in different ways, depending on the model
16   and the circumstances.  In some cases, the TK model may be used directly to perform
17   interspecies extrapolation (i.e., to derive a Human Equivalent Concentration or Dose that
18   includes TK considerations), thus alleviating the need for £FAK- In other cases, the TK model
19   may be used to derive £FAK.
20
21   3.2.2.2.  Use of Ratios
22          When AUC or concentration at steady state is the relevant dose metric, and if advanced
23   TK models are not available, £FAK can be derived using a ratio of doses producing the same
24   AUC value.  This is accomplished by identifying external doses associated with the AUC value
25   produced in animals at the point of departure (AUCA), demonstrated below (see Equation 1 and
26   Figure 4). The human dose that produces the same AUC value as  observed in animals (AUCA) is
27   the TK equivalent dose. In these cases, differences between the animal and the human dose
28   producing the same AUC value in each species are captured as the DDEF. See below:
29
30
31
32
                This document is a draft for review purposes only and does not constitute Agency policy.
     5/11/11                              Page 25 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
 1                                    AUC = Dose/Clearance                              (1)
 2
 3
 4
 5
 6   Using these data, £FAK is calculated according to Equation 2.
                                      FF  = -
                                      tLtAK
                                               fH                                  (2)

 9
10
11          Using AUC as an example, the value for the dose metric would be AUC A, that is the
12   AUC value determined in animals at the POD. Thus, in this example, Equation 2 can be

13   conceptualized as
14

15                       EFAK = DA producing AUC A / DH producing AUCA                   (3)
16

17   where
18          AUCA  = area under the curve in the animal

19          DA      = the animal external dose

20          DH      = the human external dose.

21
22
23          This is graphically presented in Figure 4.
24
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                              Page 26 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
 1
 2
 3
 4
 5
 6
 7
 8
 9
10
11
12
13
14
15
            o
            <
                                   Human
                                                            Animal
                                     DH
                                         DOSE
            Figure 4. Interspecies extrapolation based on AUC. In this example, AUC is the
            appropriate dose metric, and the relationship between dose and AUC is determined in
            animals and in humans. This method can be used to develop a Human Equivalent Dose
            or Concentration, or in the calculation of £FAK. Calculation of £FAK requires knowledge
            of applied doses in animals (DA) and in humans (DH) that produce the AUC value
            determined in animals at the point of departure (AUCA).
      Because clearance values are the mathematical reciprocal of internal dose (AUC), they
may also be used to calculate a DDEF value. As humans are assumed to be more sensitive than
experimental animals (in this instance, they clear toxicants more slowly), the human clearance
value remains in the denominator. Calculations using Cmax are developed in a manner similar to
that for AUC.

3.2.3.  Relationship to Other EPA Guidance
      The development and use of advanced information on tissue dosimetry to serve as the
basis for quantitative, nondefault uncertainty factors is consistent with existing EPA guidance
and philosophy (U.S. EPA, 2006, 2002).  The EPA's Inhalation Reference Concentration
Methodology presents a continuum of approaches from complete lack of knowledge to
     5/11/11
          This document is a draft for review purposes only and does not constitute Agency policy.
                                    Page 27 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
 1    biologically based dose-response models (U.S. EPA, 1994). There are two approaches that each
 2    cover the interval between lack of knowledge and knowledge of target tissue and target tissue
 3    concentrations in test species and humans. The first is for reactive (Category 1) gases and
 4    inhaled particles that damage portal of entry (respiratory tract) tissues, and the second is for
 5    gases that are absorbed and produce their toxicity in tissues bathed by circulating blood
 6    (Category 3 gases). For Category 1 gases, toxicity information identifies the affected region of
 7    the respiratory tract and species differences in regional respiratory tract surface area and airflow
 8    (respiratory rate) serve as the basis to quantify species differences in dosimetry.  For Category 3
 9    gases, species differences in the solubility of the compound in blood (the blood:air partition
10    coefficient) serve as the basis upon which to  quantify species differences in dosimetry. More
11    recent guidance (U.S. EPA, 2011) on orally encountered toxicants instructs the application of
12    dose scaling by the ratio of species body weights raised  to the 3/4 power (human BW:animal
13    BW)3 4.  Each of these guidance documents indicates that their approaches are intermediates, to
14    be superceded when more detailed information on tissue dosimetry can be developed. The
15    subj ect of the present guidance is the development and interpretation of quantitative
16    toxicokinetic data for the purpose of developing nondefault values for inter- and intraspecies
17    uncertainty/extrapolation.
18
19    3.2.4. Conclusions for EFAK
20          Mathematically, EFAK may be the ratio of the external doses in animals at or near the
21    POD to the value at a central tendency measure of external doses in the general human
22    population that each result in the same level of the dose  metric (this represents developing the
23    Human Equivalent Concentration or the Human Equivalent Dose). £FAK values may be
24    calculated for multiple organs/effects.  The same PODs  should be used for test animals and
25    humans for a given organ/effect. Quantitatively, DDEF values for UFA components might be
26    less than  1 if humans are less sensitive.
27          Confidence in EFAK is increased when decisions and calculations are well documented
28    and made transparent.  This includes description of toxicity data identifying the target tissue,
29    chemical species, MO A, and species concordance of effects.  Data describing the TK, the
30    metabolism of the compound, and the relationship between administered dose and internal dose

                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                               Page 28 of 49    DRAFT: DO NOT CITE  OR QUOTE

-------
 1    are also summarized.  Data that show a linear relationship between external dose and internal
 2    dose metrics can be specifically reiterated in this description; this will indicate generalizability of
 3    the EFAK to doses that may be higher or lower than those used in its calculation.  Because
 4    animal-to-human differences in target tissue concentrations may not be consistent for all
 5    responding tissues or organs, a comparison of POD and DDEF values from multiple affected
 6    organs will increase confidence in health-protectiveness of the developed DDEF value for the
 7    critical organ. Results can be presented in tabular form for ease of comparison across endpoints.
 8
 9    3.3.  INTRASPECIES TOXICOKINETIC EXTRAPOLATION (EFHK)
10          This section provides a discussion of the quantitative differences in the TK among
11    humans for intraspecies extrapolation (EFmc). From a toxicokinetic standpoint, among humans
12    experiencing the same external dose, susceptibility is due to higher target tissue concentrations of
13    the toxicant in some individuals or groups relative to the  majority of the human population. TK
14    differences for intraspecies extrapolation are characterized as the ratio of an internal dose metric
15    attained in the general human population to the dose metric from susceptible humans exposed to
16    the same external dose or concentration.  Illustrative examples are included in the appendix.
17
18    3.3.1.  Considerations  for EFmc
19    3.3.1.1.  Susceptible Groups or Individuals
20          Potentially susceptible groups/lifestages (e.g., children, elderly, or age-related
21    susceptibility, or those with a disease making them susceptible to a toxicant) can be identified.
22    In some instances, individuals may have a condition or difference in some physiologic or
23    biochemical process that may be a deterministic factor in dosimetry that serves as the basis for
24    assumed sensitivity (e.g., polymorphism of a given metabolizing enzyme). In these cases, the
25    distribution of the dose metric among the population will not have a unimodal distribution (see
26    Figure 5, left panel). In other cases, sensitivity may be distributed throughout the general
27    population, and  sensitive individuals may be those in the tail of the distribution (i.e., unimodal;
28    see Figure 5, right panel).  Documenting this information and/or these assumptions serves as the
29    basis for selecting a unimodal or a bimodal distribution of sensitivity.  The selection of a bimodal
30    or unimodal-based analysis will be a function of the available data and must be made on a

                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                               Page 29 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
     uj
     **"*
     <•>
     UJ
1
2
3

4

5

6

7
          o

          0.

          yj
                                                                              General
                                                                -OR-
                   u.

            Figure 5. Intraspecies toxicokinetics. From a toxicokinetic standpoint, susceptibility is
            based on attaining higher target tissue concentrations of the toxicant. For this evaluation,
            a fixed exposure is studied, and human interindividual variability is measured as
            differences in the value of the dose metric between sensitive and generally representative
            population groups. When a group of the population can be identified as potentially
            susceptible, toxicokinetic data from that group can be compared to the general population
            (left panel). EFmc should be  determined as the ratio of the level of the dose metric at
            some point in the upper 50% of the distribution (i.e., 95th, 97.5th, 99th percentile) for those
            deemed sensitive to the level of the dose metric at a central tendency measure of the
            general population. However, when a separate distribution of values for the susceptible
            population is not available, then a slightly different analysis is conducted (right panel).  In
            this case, EFHK should be determined as the ratio of the level of the dose metric  at a
            percentile of the population considered sensitive (i.e., 95th, 97.5th,  99th percentile) to the
            level of the dose metric at a central tendency measure of the general population.
chemical-by-chemical basis.  The decision to quantify differences via a unimodal or bimodal

analysis should be undertaken on a chemical-by-chemical basis and be based on the strength of

the available data for the chemical and for the susceptible population group. One of the

challenges in conducting a bimodal analysis will be the availability of data describing chemical

disposition and/or physiological and biochemical parameters for the susceptible population

group.
     5/11/11
           This document is a draft for review purposes only and does not constitute Agency policy.
                                       Page 30 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
 1    3.3.1.2.  Target Tissues
 2          When responses are observed in several organs at or near the same dose level, a
 3    comparison of EFHK values developed for those tissues will be informative. However, the
 4    selection of a target organ for calculating human variability other than the one serving as the
 5    basis for animal-to-human extrapolation needs to be accompanied with a justification.  This also
 6    extends to instances in which human studies identify effects in an organ or tissue and variability
 7    is assessed in another organ or tissue.  The extrapolation approach will be perceived as most
 8    consistent when the same tissue or organ is used for each phase of the extrapolation procedure.
 9
10    3.3.1.3.  Dose Response
11          Because variability in internal dosimetry may be a function of dose, the  selection of the
12    external exposure (e.g., inhaled concentration or orally ingested dose) is important.
13    Conceptually, the animal POD is first extrapolated to produce a toxicokinetically equivalent
14    human dose or concentration. The DDEF for interspecies differences in TD should be applied to
15    the tissue concentration defining the Human Equivalent Exposure to complete interspecies
16    adjustment. It is this species-adjusted concentration that is most appropriate for application in
17    the evaluation of human interindividual variability.
18          The basis for comparison of human variability is at the level of the internal, rather than
19    the external dose. Thus, it is  important that the relationship between internal and external doses
20    be well characterized.  Comparisons of the external doses that produce the same level of the
21    internal dose (between susceptible and nonsusceptible groups) can be substantially complicated
22    when there are nonlinearities in the relationship between external dose and internal
23    concentration. This is particularly difficult when nonlinearity of dose is for one group and not
24    for the other.  This complication can be avoided when the comparison is made because
25    differences in internal doses developed from the same external exposure.
26          Specific differences among humans, particularly those demonstrated in vitro (i.e.,
27    intrinsic clearance), are most reliable when they are translated into differences in dosimetry.
28    This may include evaluations of multiple different doses.
29
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                                Page 31 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
 1    3.3.2. Computation
 2          For intraspecies TK extrapolation, differences in dosimetry are characterized for human
 3    groups or lifestages by comparison to the general population.  Thus, the comparisons are among
 4    differences in internal dosimetry (or target site dose) resulting from the same external exposure.
 5    To address human variability, some attention must be devoted to identifying a susceptible
 6    population group or a fraction of the general population considered to be sensitive. TK data are
 7    then analyzed to determine values for the dose metric in the general and sensitive groups.
 8
 9    3.3.2.1.  Use of TK Models
10          A PBPK or other TK model provides the most robust approach for evaluating
11    intraspecies TK extrapolation.  The model is subject to  evaluation as previously described
12    (U.S. EPA, 2006). When an appropriate model is available, it can be used in different ways
13    depending on the model. In some cases, the TK model  may directly account for within human
14    variation and/or include data from the sensitive group, thus eliminating the need for £FHK-  In
15    other cases, the TK model may be used to derive dose metric values for calculating the value of
16    EFHK.
17
18    3.3.2.2.  Use of Ratios
19          Where TK models are not available, EFnK can be derived using a ratio (see Equation 4).
20    The value for the dose metric employed for the general  population (e.g., AUCgen) would be that
21    level  of the dose metric identified from initial studies with animals or humans and further
22    extrapolated to account for interspecies differences in toxicodynamics. The  value for the dose
23    metric in sensitive individuals (e.g., AUCsens) should be determined from empirical data or
24    pharmacokinetic modeling.
25
26
                                         "^   AUCgenor<
28
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                               Page 32 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
 1    where
 2       EFnK    = factor for intraspecies extrapolation covering toxicokinetics
 3       AUCgen  = area under the curve at a measure of central tendency in the general human
 4                  population
 5       AUCsens  = area under the curve at a percentile of interest in the sensitive human population
 6       Clgen     = clearance at a measure of central tendency in the general human population
 7       ClSens    = clearance at a percentile of interest in the sensitive human population.
 8
 9
10          Because clearance values are the mathematical reciprocal of internal dose (AUC), they
11    may also be used to calculate a DDEF value. Because susceptible humans may be assumed to be
12    more sensitive than general humans (e.g., they clear toxicants more slowly), the sensitive human
13    clearance value remains in the denominator.
14
15    3.3.3.  Conclusions for EFnK
16          The EFnKis essentially a comparison of internal doses resulting from the same external
17    exposure to both the general population and sensitive individuals.  When using an empirical
18    ratio, the £FHK is the ratio of the dose metric value at a percentile of the distribution intended to
19    represent sensitive individuals to the dose metric value at a central tendency measure of the
20    general population. Quantitatively, DDEF values for human interindividual variability cannot be
21    less than one.
22          The dose selected for quantifying human interindividual variance may have an impact on
23    the magnitude of variability. The dose adjusted from the animal POD is the preferred dose for
24    quantitation of human variability. While DDEF values may be calculated for multiple
25    organs/effects (or even doses), the same external dose needs to be used as the starting point when
26    calculating dose metric values for the sensitive and the general populations.
27          Documentation should describe the mathematical method to be employed, the type of
28    distribution and percentile(s) of interest; the dose metrics (e.g., AUC of parent compound in
29    kidney), and the target tissues for which EFHKare developed.  Confidence in the extrapolation is
30    improved when distribution types are justified or explained.
31
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                               Page 33 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
 1                       4. DDEFs BASED ON TOXICODYNAMICS (TD)

 2    4.1.  GENERAL CONSIDERATIONS
 3           Toxicodynamics describes the critical interaction of the active chemical moiety with the
 4    target site and the ensuing sequence of events leading to toxicity. Data that describe the dose-
 5    response relationship serve as the basis for deriving extrapolation factors for toxicodynamic
 6    components. This section provides a discussion of factors common to inter- and intraspecies
 7    extrapolation for TD.  In contrast to TK, which focuses on differences in internal dosimetry, TD
 8    differences are quantified as differences in concentration6 producing the same level of response
 9    between animals and humans, as well as among humans.  TD evaluations may include multiple
10    response levels, critical effects, key events, or analytical methods.  Developing aDDEF  for TD
11    requires knowledge about the relationship between an event measured  in vitro or in vivo (e.g.,
12    receptor binding) and the end result (the adverse health condition). Doses or concentrations
13    producing the measured event can be obtained in vitro or  in vivo, but they must be obtained in
14    the tissue of interest or a suitable  surrogate.  TD models, the most complex of which may be
15    represented by BBDR models, represent an important tool through which predictions may be
16    confidently made to extend the range of empirical observations. Comparisons between animals
17    and humans or to quantify human interindividual differences are made on the basis of doses or
18    concentrations that produce the same level of the same measured response.
19           Quantitative differences between animals and humans are used for interspecies
20    extrapolation (£FAD), whereas differences in susceptibility within the human population are used
21    for the intraspecies extrapolation  (EFno). Thus, the £FAD is used to extrapolate findings in
22    laboratory animals to the general  human population, and the EFHois used to extrapolate  to
23    sensitive human groups and life stages. The TD portion of each (£FAD, EFno) is combined  with
24    the corresponding TK factor to form the composite extrapolation factor.
25           Although a complete MOA understanding is not required, derivation of a DDEF  for TD
26    relies heavily on understanding a  MOA for the critical effect(s) identified for risk assessment.
27    Important questions to address include the following:
      6 When using a biologically based dose-response model, differences may be quantified as the ratio of dose metrics in
      respective species or population groups producing the same level of the response.  However, DDEF values for TD
      may also be quantified as the ratio of in vitro concentrations producing the same level of the response.
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                                Page 34 of 49    DRAFT:  DO NOT CITE OR QUOTE

-------
 1       •  What is/are the critical effect(s) and POD(s) being used for this assessment? (Each of
 2          these should be considered in this process.)
 3       •  What is the MOA or mechanism(s) for that toxicity?  Have the key events been
 4          identified? Can they be measured?
 5       •  Is the MOA or mechanism(s) in the animal model relevant to humans (qualitatively
 6          and/or quantitatively)?
 7       •  Are the data on the key events amenable to modeling such that a uniform measure in
 8          animals and humans can be derived?  If not, do the available data points include a
 9          response level that is sufficiently similar in animals and humans?
10       •  Are there data in human populations that describe population variation? Do these data
11          identify a sensitive group(s) or lifestages? Can the degree of this sensitivity be
12          estimated?
13
14
15    4.1.1. Mode of Action
16          TD extrapolation should be endpoint driven (considered in the context of the toxic
17    endpoints most relevant for purposes of risk assessment). The choice of critical effect should be
18    justified, when possible, based on findings of response in exposed humans. Understanding MOA
19    for the agent(s) of interest helps to ensure that the TD responses used  to derive the DDEF will be
20    robust scientifically.  These responses  could include receptor affinity, enzyme inhibition, and
21    molecular changes, among others.  Repair of DNA or tissue damage, biological thresholds,
22    residual function, and other processes that could contribute to nonlinearity in effect are
23    considered and discussed.  Experimental systems and measured responses should be the  same or
24    comparable.
25
26    4.1.2. Relating Response to Dose or Concentration
27          TD data may be developed from the results of in vivo or in vitro studies.  In some cases,
28    in vivo data may be used, but care should be taken to control for the influence of toxicokinetics.
29    If there are existing human data measuring the response, then these data can be used to derive the
30    POD, thereby removing the need for the UFA. Likewise, if there are data from the most  sensitive
31    human group, those data could be used for the risk assessment, and an UFn may not be needed.
32          Care should be taken to ensure that kinetic factors do not confound the interpretation  of
33    response data.  Comparisons based on  internal dose metric are preferred over applied dose; that
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                               Page 35 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
 1    is, the concentration producing the level of response (rather than applied dose or concentration)
 2    is preferred so that TD response may be distinguished from TK differences. Tissue-specific
 3    metabolism could also influence the actual target concentrations and must be considered. Blood
 4    levels are an acceptable measure of internal dose when it can be shown that they are proportional
 5    to concentrations of toxicant in target tissue or biological preparation.
 6          When using in vitro systems, the response measured should be representative of the
 7    toxicity; that is, the measured endpoint should be the same as—or highly related to—the critical
 8    effect or key event. In cases where the measured response differs, it is important to describe the
 9    potential impact of these differences on the final DDEF.  When using in vitro data, the
10    comparability of chemical uptake  between animal and human tissues should be demonstrated.  In
11    vitro results should include data describing the test chemical metabolism by the system used to
12    generate response data.
13
14    4.1.3. Range of Doses or Concentrations
15          The relationship between the doses, tissue concentrations, and/or in vitro concentrations
16    used to derive the DDEF, and those attained in the toxicity studies characterizing the POD,
17    should be characterized.  Optimally, the concentrations used in studies characterizing response
18    include the concentration at the POD.  This  is important because the variability in the response
19    may change with increasing or decreasing dose or concentration. Doses  and/or tissue
20    concentrations may also be compared to those expected from environmental exposure; this is
21    part of the consideration of overall relevance of the  test system.
22
23    4.2.  INTERSPECIES TOXICODYNAMIC EXTRAPOLATION (EFAD)
24          This section provides information on calculating TD differences between species.  These
25    are characterized as the ratio of the concentrations (concentrations used in vitro or the level of
26    the dose metric) in animals and humans producing the same level of response. Note that the
27    level of comparison is the response, and it is not a comparison of responses produced by the
28    same concentration (see Figure 6). Illustrative examples are included in  the appendix.
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                               Page 36 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
 i
 2
 3
 4
 5
 6
 7
 8
 9
10
11
12
13
14
15
16
                               Toxicodynamics
  w2
  t£  <
  iy  ^
       UJ
                                 CONCENTRATION
      Figure 6. Interspecies toxicodynamics. The TD difference between test animals and
      humans is calculated from dose or concentration-response relationships most often
      determined in test systems or animals generally representative of the respective species.
      The comparison is made for doses or concentrations producing the same level of
      response, and comparisons are made using values representing the central tendency.
4.2.1.  Considerations for
4.2.1.1. Mode of Action
      Endpoint(s) must be chosen from which to derive the £FAD, and it is preferred for the
endpoint(s) to be the actual critical effect or a key event in the TD pathway. For each endpoint
evaluated, it must be determined whether the data are from an animal typical of the responding
species/strain/sex/lifestage opposed to an animal model system that may be unusually sensitive
for a particular effect.  This might be important in transgenic animals and in animal models used
to study specific human diseases (e.g., spontaneously hypertensive rats). Animal models and
MOA(s) based on them are evaluated for human relevance.

4.2.1.2. Target Tissues
      Where there are data from the molecular targets in both species, but the data are not from
the critical target organ, the data can be used only if there is sufficient information that one tissue
is an appropriate surrogate for another. For example, a target enzyme may be present in several
5/11/11
               This document is a draft for review purposes only and does not constitute Agency policy.
                                       Page 37 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
 1    tissues, including blood; however, blood is easily obtained from humans, but tissue from the
 2    target organ is not. Studies may show that the chemical effect on this target enzyme in blood
 3    correlates very well and is entirely predictive of the effect in the target organ, and indeed the
 4    enzyme structure, function, and chemical affinity is the same regardless of tissue. Such
 5    information would be necessary to allow using comparisons of chemical effects in the blood
 6    enzyme in both humans and laboratory animals.
 7
 8    4.2.1.3. Dose Response
 9           The choice of response level to use for comparison depends on many factors:
10
11       •   Completeness of dose response
12       •  Linearity of the dose response
13       •  Understanding of the effect along the toxic pathway at that response level
14
15
16           The magnitude of £FAD may be a function of the response level chosen for extrapolation.
17    Confidence is increased when the response level employed for £FAD calculation approximates
18    the response level at the POD and when the ratio of doses producing the same response level in
19    animals and humans is similar over a range of doses. When data are available to describe the full
20    dose-response curve, evaluating the shape of the dose curves for animals and humans can
21    provide important information. If the shapes of the curves are  different, then the magnitude of
22    £FAD will depend on the response level selected. Note that concentrations causing greater
23    magnitudes of effect may also produce nonspecific cellular changes that could confound the
24    comparisons.
25
26    4.2.1.4. In Vitro Data
27          When using data from in vitro systems, the activity of the parent chemical and/or
28    metabolites, as well as the extent of metabolism of the compound by the in vitro system, should
29    be known. The in vitro assay should measure a response that can be linked to the toxic outcome,
30    and the assays should employ the same (responding) tissues from the in vivo test animal
31    species/strain/sex/lifestage and from humans. Experimental systems should be as closely
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                               Page 38 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
 1    matched between species as possible, and the concentrations of toxicant in vitro should be
 2    compared to tissue concentrations (the dose metric) at the POD.  See Section 2.2.3.2 for other
 3    general considerations.
 4
 5    4.2.2. Computation
 6    4.2.2.1.  Use of TD Models
 1          A biologically based dose response or other TD model provides the most robust approach
 8    for evaluating interspecies TD extrapolation. The model is subject to evaluation as described
 9    previously (U.S. EPA, 2006).  When available, the model can be used to inform several
10    decisions, depending on the model.  In some cases, the TD model may be used directly to
11    perform interspecies extrapolation and to derive a Human Equivalent Concentration or Dose that
12    includes TD considerations, thus alleviating the need for £FAD.  In other cases, the TD may be
13    used to derive the £FAD used.
14
15    4.2.2.2.  Use of Ratios
16          When PD models are not available, EFAo would be calculated as a ratio describing the
17    relationship between the tissue concentrations producing a set response in human compared to
18    animal tissues, preferably at, or near, the response level at the POD. For example, "x response
19    level" may be a 10% response if EDio or BMDio is used. The £FAD would be calculated as
20    follows:
21
                                 ^  _ ConcentrationA.xresponselevel
22                              &TAD - n       ,  ,•                                         (•))
                                       ConcentrationH
-------
 1          To evaluate the extent to which the shape of the dose-response curve varies between
 2    animals and humans and, thus, impacts the magnitude of the £FAD, a range of response levels
 3    should be evaluated. The rationale and implications for choosing the point for extrapolation
 4    should also be presented.
 5
 6    4.2.3.  Conclusions for £FAD
 7          Mathematically, the £FAD will be the relationship between the concentrations or dose
 8    metric values resulting in the same level of response in  both the test species and the generally
 9    representative human (often the 70-kg adult male). DDEF values may be calculated for multiple
10    PODs and organs/effects, but the same response levels in animals and humans should be used for
11    quantitation when possible. Confidence in the value (knowing whether the DDEF value may
12    vary depending on the response level) is improved when EFAD values developed from multiple
13    points on the concentration-response curve are compared; the shape/slope of the curves may also
14    influence these values.  Quantitatively, £FAD can be less than one if the data show humans are
15    inherently less sensitive than animals.
16          A summary of all conclusions and their scientific support should be provided.  Data
17    describing the dose response of the compound in animals and humans can be specifically
18    reiterated in this description; this will indicate the extent to which the £FAD value can be
19    generalized to doses that may be higher or lower than those used in its calculation.
20
21    4.3.  INTRASPECIES TOXICODYNAMIC EXTRAPOLATION (EFHD)
22          This section describes derivation of the EFHD- TD variability within the human
23    population is calculated as the relationship between concentrations or dose metric values
24    producing the same level of the response in the general  population and in susceptible groups or
25    individuals.  From a toxicodynamic standpoint, susceptibility is based on attaining a given level
26    of response at a lower concentration of toxicant.  For this evaluation, multiple response levels,
27    critical effects (or key events), analytical methods, or susceptible groups or individuals may be
28    considered.  No data sets were identified upon which a  conclusive case study example could be
29    developed for intraspecies toxicodynamic extrapolation.
30

                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                               Page 40 of 49  DRAFT: DO NOT CITE OR QUOTE

-------
 1    4.3.1.  Considerations for
 2    4.3.1.1. Susceptible Groups or Individuals
 3           Susceptibility in the human population may be due to lifestage, health status or disease
 4    state, genetic disposition, or other factors.  Considering susceptibility to more than one critical
 5    effect may require consideration of more than one life-stage; critical windows of development,
 6    and, therefore, windows of susceptibility, occur at different times for various tissues, organs, and
 7    systems.  Currently, sufficient data to address susceptibility  are rarely available; however,
 8    research in this area is rapidly expanding. For example, population variation, such as genetic
 9    polymorphisms, is an expanding area of study.  It is anticipated that the increased availability
10    and experience applying "omics" technologies will benefit the derivation of DDEFs, in general,
11    and EFno, in particular. A data-derived EFnois feasible, given human data are of sufficient
12    quality; the data address aspects of the critical effect consistent with that identified from
13    applicable human or animal studies; and the studies have been conducted in the segment(s) of
14    individuals  or the population deemed sensitive.
15           Ideally, data will be robust enough to enable more than point estimates in the general and
16    susceptible  groups.  As discussed  in more detail below, distributional analysis of response  data
17    should be conducted to identify points for use in quantitation. The relationship between the
18    measured response and the toxicity endpoint of concern (e.g., critical effect or key event) should
19    be described, whether determined in vivo or in  vitro.
20
21    4.3.1.2. Target Tissues
22           For  calculation of EFno, data for multiple responding tissues can be evaluated, and
23    multiple DDEFs can be derived.  It is particularly important to evaluate those tissues that
24    demonstrate response at doses or concentrations near those for the critical effect.
25
26    4.3.1.3. In Vitro Data
27           Given the constraints on generation of human response data in vivo, in vitro  studies offer
28    an appealing alternative.  Samples selected for in vitro investigation should represent the general
29    human population as well as those groups or individuals thought or demonstrated to be
30    susceptible.  See Section 2.2.3.2 for other general considerations.
31
                 This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                               Page  41  of 49   DRAFT: DO NOT CITE OR QUOTE

-------
 1    4.3.2.  Computation
 2           For TD extrapolation, the goal is to determine the difference between humans on the
 3    basis of concentration producing the same response level. For quantitation, data on the critical
 4    response(s) are derived from a population that includes susceptible groups or individuals.
 5    Because the data available to define potentially susceptible groups or individuals could be
 6    viewed in different ways, a statistical analysis may be helpful to determine distribution type (see
 7    Figure 7):
 8
 9       •   A unimodal distribution where the potentially susceptible group(s) represent the tail of
10           the distribution because they cannot be separated from the general population.
11       •   A bimodal (or multimodal) distribution where the group(s) can be readily identified.
12
13
14           Documenting critical response data, assumptions made, and the distribution selected will
15    serve as the basis for quantitation.
16
17    4.3.2.1. Use of TD Models
18           A biologically based dose response or other TD model provides the most robust approach
19    for evaluating intraspecies TD extrapolation.  When sufficient data are available, these TD
20    models can be structured and exercised to include differences in mode-of-action components that
21    may be lifestage-dependent or influenced by other potentially susceptibility-inducing conditions
22    such as genetic polymorphisms. Specific to EFno, it is critical that the model parameter
23    reflecting the underlying cause of susceptibility in a group be well documented.  When an
24    appropriate model is available, it can be used in different ways depending on the model. In some
25    cases, the TD model may directly account for within human variation and/or include data from
26    the sensitive group, thus eliminating the need for EFno-  In other cases, the TD may be used to
27    derive EFno-
28
29    4.3.2.2. Use of Ratios
30           When TD models are not available and there are groups or individuals that can be
31    identified as sensitive, then the EFno may be defined as the ratio between the concentrations
32

                 This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                               Page 42 of 49  DRAFT: DO NOT CITE OR QUOTE

-------
(0
UJ

     yj    «
                                                   -OR-
                                                   -OR-
                                                                B1
                                                                             General

                                                                B2

   Figure 7. Intraspecies toxicodynamics. DDEF values for TD are defined by ratios of
   concentrations producing the same level of response in the general population and some
   defined percentile (e.g., 1st, 2.5th, 5th, etc.) for the distribution representing sensitive
   individuals. When a specific group of the population can be identified as potentially
   susceptible, TD data from that group can be compared to the general population (Panels Al
   and A2). Panel Al presents a dose-response curve (cumulative distribution plot) for both
   populations that demonstrates the central tendency (solid line) and confidence bounds, or
   bounds of variability (dashed lines), for data obtained from the general population and from
   an identifiable sensitive group.  In this example, the level of response (Y-axis) has been
   selected (e.g., 10%-response level), and the concentrations producing this level of response in
   the general and sensitive populations/groups are obtained from the X-axis. Panel A2 is
   derived from the same data used for Panel Al, but it presents the distribution of
   concentrations producing the defined level of response only; no other dose-response data are
   carried over into Panel A2.  Alternately, when potentially susceptible individuals represent a
   small percentage of the general population (Panels Bl and B2), a slightly different analysis is
   conducted.  In this case, EFno should be determined as the ratio of the concentrations
   producing the same level of response (1) at a measure of the central tendency in the
   population to (2) the concentration producing the response level at a percentile of the general
   population considered sensitive (e.g., 1st, 2.5th, 5th percentile).  Panel Bl demonstrates this
   comparison using the concept of confidence bounds on the dose-response relationship, and
   Panel B2 demonstrates the distribution of concentrations producing the response, only at the
   response level chosen for comparison (e.g., the 10%-response level).
           This document is a draft for review purposes only and does not constitute Agency policy.
5/11/11                               Page 43 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
 1    producing the same level of response in the general population and a lower percentile in the
 2    sensitive group (see Figure 7) using Equation 6:
 3
                                             Concentration pn
 4                                   EFHD =	s—                                (6)
                                             Concentrationsens
 5
 6    where
 7           EFHD               = factor for intraspecies extrapolation covering toxicodynamics
 8           Concentrationgen     = concentration producing the in response the general human
 9                                 population
10           ConcentrationSens     = concentration producing the response at a percentile of interest for
11                                 the sensitive group.
12
13
14           When sensitivity among the population exhibits a unimodal distribution, the EFno is the
15    ratio of the concentration that elicits a level of response at the central tendency of the distribution
16    to the concentration that elicits the same level of response in sensitive individuals (e.g., 5*, 2.5*,
17    and 1st percentiles of the distribution; sensitive individuals will respond at lower concentrations).
18    It is important to define and justify the point(s) in the distribution representing sensitive groups
19    or individuals.
20           When sensitivity among the population exhibits a bimodal (or multimodal)  distribution,
21    the DDEF is determined in a similar manner, using the concentrations (e.g., 5th, 2.5th, and 1st
22    percentiles of the concentration distribution) that elicit the specific level of response in the
23    sensitive individuals for the most susceptible group(s).  The values selected to describe the
24    potentially sensitive group(s) or individuals are defined and presented at varying levels.  The
25    selection of the response level  and the percentile of the distribution used to describe the
26    potentially sensitive group(s) or individual(s) is an important issues.  This is a situation where the
27    communication between risk assessment and risk management is essential.
28
29    4.3.3.  Conclusions for EFHo
30           A biologically based dose response or other TD model provides the most robust approach
31    for evaluating intraspecies TD extrapolation. When using empirical ratios, the EFno will be the
                 This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                                Page 44 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
 1    ratio of the concentration producing the specified level of response in the general human
 2    population to the concentration producing the same level of response in susceptible groups or
 3    individuals.  Increased confidence in the EFHo is developed when the concentration used for the
 4    comparison of responses is compared to doses or concentrations at the POD. Quantitatively,
 5    EFno cannot be less than one.
 6          The risk assessor describes all choices and their rationales, including the use of multiple
 7    response levels, critical effects (or key events), analytical methods, or data from susceptible
 8    groups or individuals.  The conclusions include a clearly worded description of the mathematical
 9    method(s) employed and a presentation of the relationship between the measured response and
10    toxicity (i.e., critical effects or key events). This description should clearly identify and provide
11    the justification for available data and points in the distribution(s) representing sensitive
12    individuals.  Attention should be paid to characterizing the distribution type employed for
13    analysis; uncertainty in the choice of distribution type can be reduced by presenting DDEF
14    values resulting from multiple distribution types.
15
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                                Page 45 of 49   DRAFT: DO NOT CITE OR QUOTE

-------
 1                                       5.  FINAL STEPS

 2          The composite DDEF is calculated after the appropriate DDEF values for inter- and
 3    intraspecies differences in TK and TD have been derived. The composite factor is calculated by
 4    multiplying the specific factors (default and/or DDEFs), as shown in Equation 7. This is entirely
 5    analogous to calculating composite UFs when using the 10* defaults for UFA and UFn. The
 6    composite DDEF may be less or greater than 100.
 7
 8
 9                               CF = EFAK x EFAD x EFHK x EFHD                           (7)
10
11    where
12          CF     = composite uncertainty factor
13          EFAK   = factor for interspecies extrapolation covering toxicokinetics
14          EFAD   = factor for interspecies extrapolation covering toxicodynamics
15          £FHK   = factor for intraspecies extrapolation covering toxicokinetics
16          EFno   = factor for intraspecies extrapolation covering toxicodynamics
17
18
19          In practice, data may only be available to develop a DDEF for one component of
20    extrapolation or another (e.g., data for EFAK but not EFAD)- In these cases, the remaining
21    extrapolation is done by an appropriate default procedure. As such, DDEFs and defaults (i.e.,
22    UFs) are used in combination. Often this default will be a 3 x  UF—as described in the existing
23    RfC methodology and the 3/4 body-weight procedure (U.S. EPA, 2011, 1994). When data are not
24    available to develop DDEFs for either component of interspecies or intraspecies extrapolation,
25    the 10x default value for the uncertainty factor is applied.
26          Finally, the composite factor provides the total magnitude of the factor.  The values
27    derived for each of the components and the resulting extrapolations should be clearly reported
28    and characterized. The relationship of each of these doses or concentrations to both the POD and
29    to doses or concentrations likely attained from environmental  exposures should be presented.
30
                This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                               Page 46 of 49  DRAFT: DO NOT CITE OR QUOTE

-------
                                               6.  REFERENCES
 2    Alarie, Y. (1973) Sensory irritation by airborne chemicals. CRC Crit Rev Toxicol 2:299-363.

 3    Barton, HA. (2005) Computational pharmacokinetics during developmental windows of susceptibility. J Toxicol
 4    Environ Health A 68(11-12):889-900.

 5    Bogdanffy, MS; Daston, G; Faustman, EM; et al. (2001) Harmonization of cancer and non-cancer risk assessment:
 6    Proceedings of a consensus-building workshop.  Toxicol Sci 61:18-31.

 7    Boobis, AR; Doe, JE; Heinrich-Hirsch, B; et al. (2008) IPCS framework for analyzing the relevance of a noncancer
 8    mode of action for humans. Crit Rev Toxicol 38:87-96.

 9    Boyes, WK; Bercegeay, M; Krantz, T; et al. (2005) Momentary brain concentration of trichloroethylene predicts the
10    effects on rat visual function.  Toxicol Sci 87(1): 187-96.

11    Brown, RP; Delp, MD; Lindstedt, SL; et al. (1997) Physiological parameter values for physiologically based
12    pharmacokinetic models. Toxicol Ind Health 13(4):407-484.

13    Carlile, DJ; Zomorodi, K; Houston, JB.  (1997) Scaling factors to relate drag metabolic clearance in hepatic
14    microsomes, isolated hepatocytes, and the intact liver: studies with induced livers involving diazepam. Drag Metab
15    Dispos25(8):903-911.

16    Houston, JB; Carlile, DJ. (1997) Prediction of hepatic clearance from microsomes, hepatocytes, and liver slices.
17    Drag Metab Rev 29(4):891-922.

18    IPCS (International Programme on Chemical Safety).  (2005) Chemical-specific adjustment factors for interspecies
19    differences and human variability: guidance document for use of data in dose/concentration-response assessment.
20    Harmonization Project Document No. 2. Geneva, Switzerland: World Health Organization.

21    Krishnan, K; Andersen, ME. (1994) Physiologically-based pharmacokinetic modeling in toxicology.  In: Hayes, WA
22    ed. Principles  and Methods of Toxicology. 3rd edition. New York, NY: Raven Press; pp.  193-242.

23    Maronpot, RR; Flake, G; Huff, J. (2004) Relevance of animal carcinogenesis findings to human cancer predictions
24    and prevention. Toxicol Pathol 32(Suppl. l):40-48.

25    Meek, ME. (2001) Categorical default uncertainty factors: interspecies variation and adequacy of database. Human
26    Ecol Risk Assess 7:157-163.

27    Meek, ME; Renwick, A; Ohanian, EO; et al. (2001) Guidelines for application of compound specific adjustment
28    factors (CSAF) in dose/concentration - response assessment. Comm Toxicol 7:575-590.

29    Meek, ME; Bucher, JR; Cohen, SM; et al. (2003) A framework for human relevance analysis of information on
30    carcinogenic modes of action.  CritRev Toxicol 33:591-653.

31    Murray, FJ; Andersen, ME. (2001) Data derived uncertainty factors; boric acid (BA) as a case study. Human Ecol
32    Risk Assess 7:125-138.

33    NRC (National Research Council). (1983) Risk assessment in the federal government: managing the process.
34    Washington, DC: National Academy Press.

35    NRC (National Research Council). (1994) Science and judgment in risk assessment.  Washington, DC: National
36    Academy Press.
                  This document is a draft for review purposes only and does not constitute Agency policy.
      5/11/11                                    Page 47 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
  1     O'Flaherty, EJ. (1989) Interspecies conversion of kinetically equivalent doses. Risk Anal 9(4):587-598.

  2     Proctor, DM; Gatto, NM; Hong, SJ; et al. (2007) Mode-of-action framework for evaluating the relevance of rodent
  3     forestomach tumors in cancer risk assessment. Toxicol Sci 98(2):313-326.

  4     Renwick, AG. (1993) Data-derived safety factors for the evaluation of food additives and environmental
  5     contaminants. Food Addit Contam 10(3):275-305.

  6     Renwick, AG. (1994) Toxicokenetics - pharmacokinetics in toxicology. In: Hayes, WA; ed. Principles and
  7     Methods of Toxicology.  3rd edition New York, NY: Raven Press; pp. 101-148.

  8     Renwick, AG; Lazarus, NR. (1998) Human variability and noncancer risk assessment—an analysis of the default
  9     uncertainty factor.  RegulToxicolPharmacol27:3-20.

10     Seed, J; Carney, EW; Corley, RA; et al. (2005) Overview: using mode of action and life stage information to
11     evaluate the  human relevance of animal toxicity data. Crit Rev Toxicol 35(8-9):664-672.

12     Sonich-Mullin, C; Fielder, R; Wiltse, J; et al. (2001) IPCS conceptual framework for evaluating a mode of action for
13     chemical carcinogenesis. Regul Toxicol Pharmacol 34:146-152.

14     Thomas, V. (1975) Biological-mathematical modeling of chronic toxicity. U.S. Air Force, Aerospace Medical
15     Research Laboratory, Aerospace Medical Division, Wright-Patterson Air Force Base, OH.  AMRL-TR-75-5.

16     U.S. EPA (U.S. Environmental Protection Agency). (1991) Guidelines for developmental toxicity risk assessment.
17     Risk Assessment Forum, Washington, DC; EPA/600/FR-91/001.  Available online at
18     http://www.epa.gov/ncea/raf/pdfs/devtox.pdf.

19     U.S. EPA (U.S. Environmental Protection Agency). (1993) Reference dose (RfD): description and use in health risk
20     assessments.  Background Document 1A, March 15, 1993. Available online at
21     http://www.epa.gov/ncea/iris/rfd.htm.

22     U.S. EPA (U.S. Environmental Protection Agency). (1994) Methods for derivation of inhalation reference
23     concentrations and application of inhalation dosimetry.  October.  Office of Health and Environmental Assessment,
24     Environmental Criteria and Assessment Office, Washington, DC;  EPA/600/8-90/066F.

25     U.S. EPA (U.S. Environmental Protection Agency). (1996) Guidelines for reproductive toxicity risk assessment.
26     Risk Assessment Forum, Washington, DC; 630/R-96/009. Available online at
27     http://www.epa.gov/ncea/raf/pdfs/repro51 .pdf.

28     U.S. EPA (U.S. Environmental Protection Agency). (1997a) Use of probabilistic techniques (including Monte Carlo
29     Analysis) in risk assessment. Memo from Deputy Administrator Fred Hansen, May 15, 1997.  Available online at
3 0     http://www.epa.gov/OSA/spc/pdfs/probcovr.pdf.

31     U.S. EPA (U.S. Environmental Protection Agency). (1997b) Policy for use of probabilistic analysis in risk
32     assessment at the U.S. Environmental Protection Agency.  May 15, 1997.  Available online at
3 3     http://www.epa.gov/OSA/spc/pdfs/probpol.pdf.

34     U.S. EPA (U.S. Environmental Protection Agency). (1998) Guidelines for neurotoxicity risk assessment. Risk
35     Assessment Forum, Washington, DC; 630/R-95/001F. Available  online at
3 6     http://www.epa.gov/ncea/raf/pdfs/neurotox.pdf.

37     U.S. EPA (U.S. Environmental Protection Agency). (2000) Science policy council handbook: risk characterization.
3 8     Office of Research and Development, Office of Health and Environmental Assessment, Washington, DC;
39     EPA/100/B-00/002. Available online at http://www.epa.gov/iris/backgr-d.htm.
                   This document is a draft for review purposes only and does not constitute Agency policy.
       5/11/11                                    Page 48 of 49    DRAFT: DO NOT CITE OR QUOTE

-------
  1    U.S. EPA (U.S. Environmental Protection Agency). (2001) Risk assessment guidance for superfund: volume III -
  2    part a, process for conducting probabilistic risk assessment. Office of Emergency and Remedial Response.
  3    Washington, DC; EPA-540-R-02-002. Available online at http://www.epa.gov/superfund/RAGS3A/index.htm,

  4    U.S. EPA (U.S. Environmental Protection Agency). (2002a) Guidelines for ensuring and maximizing the quality,
  5    objectivity, utility and integrity for information disseminated by the Environmental Protection Agency.  Office of
  6    Environmental Information, Washington, DC; EPA/260/R-02/008. Available online at
  7    http://www.epa.gov/quality/informationguidelines/documents/EP A_InfoQualityGuidelines.pdf.

  8    U.S. EPA (U.S. Environmental Protection Agency). (2002b) A review of the reference dose and reference
  9    concentration processes. December. Risk Assessment Forum. Washington, DC; EPA/630/P-02/002F.

10    U.S. EPA (U.S. Environmental Protection Agency). (2004) An examination of EPA risk assessment principles &
11    practices. Staff Paper Prepared for the U.S. Environmental Protection Agency by members of the Risk Assessment
12    Task Force. Office of the Science Advisor, Washington, DC; EPA/100/B-04/001.

13    U.S. EPA (U.S. Environmental Protection Agency). (2005) Guidelines for carcinogen risk assessment. Risk
14    Assessment Forum, Washington, DC; EPA/630/P-03/001F. Federal Register 70(66): 17765-17817. Available
15    online at http://www.epa.gov/raf.

16    U.S. EPA (U.S. Environmental Protection Agency). (2006) Approaches for the application of physiologically based
17    pharmacokinetic (PBPK) models and supporting data in risk assessment (final report). U.S. Environmental
18    Protection Agency, Washington, DC; EPA/600/R-05/043F.

19    U.S. EPA (U.S. Environmental Protection Agency) (2011) Recommended Use of Body Weight3/4 as the Default
20    Method in Derivation of the Oral Reference Dose. Risk Assessment Forum,  Washington, DC; EPA100/R-11/001.
21    Federal Register, 76(38): 10591-10592.  Available online at http://www.epa.gov/raf.
                   This document is a draft for review purposes only and does not constitute Agency policy.
       5/11/11                                    Page 49 of 49    DRAFT: DO NOT CITE OR QUOTE

-------