Toxicokinetic and toxicodynamic considerations when deriving health-based exposure limits for pharmaceuticals

Toxicokinetic and toxicodynamic considerations when deriving health-based exposure limits for pharmaceuticals

Accepted Manuscript Toxicokinetic and toxicodynamic considerations when deriving health-based exposure limits for pharmaceuticals John F. Reichard, M...

1MB Sizes 0 Downloads 63 Views

Accepted Manuscript Toxicokinetic and toxicodynamic considerations when deriving health-based exposure limits for pharmaceuticals John F. Reichard, M. Andrew Maier, Bruce D. Naumann, Alison M. Pecquet, Thomas Pfister, Reena Sandhu, Edward V. Sargent, Anthony J. Streeter, Patricia A. Weideman PII:

S0273-2300(16)30140-4

DOI:

10.1016/j.yrtph.2016.05.027

Reference:

YRTPH 3587

To appear in:

Regulatory Toxicology and Pharmacology

Received Date: 5 May 2016 Accepted Date: 19 May 2016

Please cite this article as: Reichard, J.F., Maier, M.A., Naumann, B.D., Pecquet, A.M., Pfister, T., Sandhu, R., Sargent, E.V., Streeter, A.J., Weideman, P.A., Toxicokinetic and toxicodynamic considerations when deriving health-based exposure limits for pharmaceuticals, Regulatory Toxicology and Pharmacology (2016), doi: 10.1016/j.yrtph.2016.05.027. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

1

Title

2

Toxicokinetic and Toxicodynamic Considerations when Deriving Health-Based Exposure Limits for

3

Pharmaceuticals

5

Authors

6

1

7

1

8

2

9

1

10

3

11

4

12

5

13

6

14

7

RI PT

4

Reichard, John F. Maier, M. Andrew

SC

Naumann, Bruce D. Pecquet, Alison M.

M AN U

Pfister, Thomas Sandhu, Reena Sargent, Edward V. Streeter, Anthony J.

Weideman, Patricia A.

15 1

Toxicology Excellence for Risk Assessment (TERA) at the University of Cincinnati

17

2

Merck & Co., Inc.

18

3

F. Hoffmann-La Roche Ltd

19

4

SafeDose Ltd

20

5

Rutgers University

21

6

22

7

EP

AC C

Johnson & Johnson

TE D

16

Genentech

1

ACCEPTED MANUSCRIPT

Contact Author

24

John F. Reichard

25

Toxicology Excellence for Risk Assessment (TERA) at the University of Cincinnati

26

Department of Environmental Health

27

College of Medicine

28

160 Panzeca Way

29

Cincinnati, OH 45267

30

[email protected]

AC C

EP

TE D

M AN U

SC

RI PT

23

2

ACCEPTED MANUSCRIPT

Abstract

32

The purpose of this paper is to describe the use of toxicokinetic (TK) and toxicodynamic (TD) data in

33

setting acceptable daily exposure (ADE) values and occupational exposure limits (OELs). Use of TK data

34

can provide a more robust exposure limit based on a rigorous evaluation of systemic internal dose.

35

Bioavailability data assist in extrapolating across different routes of exposure to be protective for multiple

36

routes of exposure. Bioaccumulation data enable extrapolation to chronic exposures when the point of

37

departure (PoD) is from a short-term critical study. Applied in the context of chemical-specific adjustment

38

factors (CSAFs), TK data partially replace traditional default adjustment factors for interspecies

39

extrapolation (extrapolation from studies conducted in animals to humans) and intraspecies variability (to

40

account for human population variability). Default adjustments of 10-fold each for interspecies and

41

intraspecies extrapolation are recommended in several guidelines, although some organization

42

recommend other values. Such default factors may overestimate variability for many APIs, while not being

43

sufficiently protective for variability with other APIs. For this reason, the use of chemical specific TK and

44

TD data is preferred. Making full use of existing TK and TD data reduces underlying uncertainties,

45

increases transparency, and ensures that resulting ADEs reflect the best available science.

46

TE D

M AN U

SC

RI PT

31

Key Words

48

Human health risk assessment, Toxicokinetic, Toxicodynamic, Pharmacokinetic, Pharmacodynamic,

49

Chemical Specific Adjustment Factor (CSAF), Bioaccumulation, Bioavailability, Cmax, Area under the

50

curve (AUC)

AC C

EP

47

3

ACCEPTED MANUSCRIPT

51

1. Introduction

52

In pharmaceutical manufacturing, acceptable daily exposure (ADE) and permitted daily exposure (PDE)

53

values are intended to protect patients against potential adverse effects from cross-contamination by

54

active pharmaceutical ingredients (APIs) that may be present in pharmaceutical products as a result of

55

the manufacturing process (EMA, 2012; ISPE, 2010; US FDA, 2008). Likewise, occupational exposure

56

limits (OELs) are used to protect workers who manufacture or process pharmaceuticals. In recent years,

57

several guidelines have advocated the use of health-based risk assessment methodologies for setting

58

ADEs and OELs (EMA, 2014; ICH, 2013, 2011; ISPE, 2010). However, these documents provide limited

59

technical guidance on how to use toxicokinetic (TK) or toxicodynamic (TD) data in the risk assessment

60

process. This contributes to differences among limits derived by different risk assessors and companies,

61

even for the same API (Snodin and McCrossen, 2012; Walsh, 2011a,b; Walsh et al., 2013). The objective

62

of this paper is to describe various approaches for using TK data in the ADE derivation process and

63

provide descriptive examples to illustrate concept application. It should be noted that these same

64

approaches can be applied for OEL derivation with specific considerations given to the inhalation route of

65

exposure, as described below.

TE D

66

M AN U

SC

RI PT

1

On a molecular level, toxic effects are not produced unless the biologically active agent (toxicant or drug)

68

reaches the appropriate target molecule(s) (such as receptors or enzymes) in the body at a concentration

69

and for a length of time sufficient to cause toxicity. The toxicity that results can be predicted on the basis

70

of two inputs: TK, which is the delivery of the drug (or its active metabolite) to the target molecule(s) (i.e.,

71

what the body does to the chemical); and TD, which is the relationship between drug concentration at the

72

target molecule(s) and the resulting adverse response, including the time course and intensity of

73

response (i.e., what the chemical does to the body). For toxicants having a non-mutagenic mode of action

74

(MOA), when the concentration of chemical is below the biological threshold of the target molecule(s)

75

there is insufficient perturbation to elicit a meaningful (i.e., clinically significant) response. In this respect,

76

consideration of available TD data is important for identifying the key TK parameters that need to be

77

evaluated in the analysis; for example, the critical effect may be a consequence of peak plasma

AC C

EP

67

1

The term ADE is synonymous with the PDE (EMA, 2014)

4

ACCEPTED MANUSCRIPT

concentrations (Cmax) or the total plasma concentration over time (AUC). The threshold below which the

79

critical effect does not occur is used as the point of departure (PoD), and demarcates the no-observed-

80

adverse-effect level (NOAEL) from the lowest-observed-adverse-effect level (LOAEL) (Bercu et al., 2016,

81

this issue). The main advantage of using TK and TD data for risk assessment is that it becomes possible

82

to integrate an internal measure of dose and response, which provides a more meaningful basis for

83

estimating risk and enables extrapolations across routes, species, and different durations of exposure.

RI PT

78

84

In toxicology and risk assessment, the terms TK and TD are often used in preference to the terms

86

pharmacokinetic (PK) and pharmacodynamic (PD). In general, PK and PD are used in relation to

87

therapeutic doses of pharmaceuticals where the intent is to provide a link between preclinical studies and

88

humans, or to characterize or tailor the therapeutic use. In contrast, TK and TD are more recent

89

extensions of the PK and PD concepts that describe adverse effects occurring at supra-therapeutic

90

doses, and carry the connotation of harm rather than therapeutic benefit (Welling, 1995). When setting an

91

ADE or OEL for a pharmaceutical, the critical effect may be one that is not considered adverse when the

92

exposure is a result of therapeutic clinical use. In patients, the potential for an adverse effect can be offset

93

by the presumed medical benefit, whereas ADEs and OELs do not reflect a presumed medical benefit

94

and, therefore, pharmacologic effects are considered adverse.

M AN U

TE D

95

SC

85

There are a range of approaches for utilizing TK/TD data in human health risk assessment; from the use

97

of bioavailability and bioaccumulation ratios to much more complex biologically-based dose-response

98

(BBDR) models and physiologically-based pharmacokinetic (PBPK) models. BBDR models are based on

99

animal-derived data that describe biological processes at the cellular and molecular level. BBDR models

100

link external exposure to an adverse apical response and estimate the probability of an adverse response

101

in humans. In a BBDR model, the linkage between exposure and adverse response is based on

102

quantifiable biological events involved in effect progression, such as apoptosis, protein induction, or cell

103

division (Crump et al., 2010). The linkage between external exposure and target site exposure (e.g.,

104

tissue concentration) can be predicted by a PBPK model. These structured, mathematical models

105

integrate in vitro, in silico, and in vivo data to evaluate the effects of various factors such as species

AC C

EP

96

5

ACCEPTED MANUSCRIPT

differences, variability within populations (e.g., age, sex, genetics, or organ dysfunction), and route of

107

administration differences on target tissue concentrations, and can be a valuable tool in predicting human

108

safety. While PBPK models are not often used for setting ADEs or OELs, their use in drug development is

109

becoming increasingly common and the resulting models can be used for risk assessment. Adaptation of

110

these advanced drug development models for risk assessment purposes lies beyond the scope of this

111

paper; however, in the absence of BBDR or PBPK models, TK/TD data can be utilized to replace default

112

uncertainties in the derivation of ADEs and OELs, as described below.

SC

113

RI PT

106

Compared to many industrial chemicals, many pharmaceutical APIs are ideally suited for use of TK/TD

115

data for risk assessment because they have rich datasets. Even in the drug discovery or development

116

phase, key TK/TD data are typically known; such as the identity of the target tissue and molecules,

117

mechanism of action, and receptor occupancy estimates. In addition, TK data on bioavailability, tissue

118

distribution, drug protein binding, metabolism, and excretion may also exist. Initially these data are

119

obtained from in vitro assays and well-designed animal studies. Later in development, TK data are also

120

available from clinical trials and post-marketing information.

TE D

121

M AN U

114

It should be noted that current guidelines for using TK and TD data for derivation of ADE limits (ICH,

123

2011; IPCS, 2005; ISPE, 2010) are predicated on a linear relationship between a dose metric (i.e., the

124

measure of a dose) and a biologic response. For the vast majority of small molecule APIs this relationship

125

is valid for exposures at and below the range of clinically-relevant doses. However, when the biologic

126

processes that regulate absorption, metabolism, and elimination are overwhelmed, the linear relationship

127

between the dose metric (i.e., AUC and Cmax, discussed below) and response is lost. Likewise, for some

128

biologic APIs, such as monoclonal antibodies, there can be nonlinearity in the sensitivity of the biologic

129

response. In either case, non-linearity in the range of clinically-relevant doses is problematic because it

130

results in unpredictable relationships between dose and response, which can include exaggerated on-

131

target pharmacology, off-target side effects, or therapeutic failure (if the API does not achieve adequate

132

systemic levels). Such non-linear effects are often observed in nonclinical animal studies, especially in

133

toxicokinetics, as dose levels increase towards maximally tolerated doses (MTD). When the relationship

AC C

EP

122

6

ACCEPTED MANUSCRIPT

between the dose metric and response lacks linearity, standard risk assessment methods for making

135

animal to human and inter-human adjustments cannot be used. However, careful evaluation of TK/TD

136

data can identify a more appropriate dose metric (Kirman et al., 2003) or a lower dosing range where

137

linearity exists. There are some APIs, such as biologics, where non-linearity exists in either the kinetics or

138

response behavior at clinically-relevant doses due to the biology associated with their MOA or kinetic

139

disposition (Muller et al., 2009). In these situations, applications of more sophisticated PBPK models may

140

be useful. One such approach is to use kinetic and dynamic data to computationally determine the

141

minimum-anticipated-biological-effect level (MABEL) from which a human equivalent dose (HED) at the

142

predicted NOEL can be obtained (Muller et al., 2009).

144

M AN U

143

SC

RI PT

134

2. Background and History: The Move from Default Adjustment Factors to Data-Derived Approaches

145

The most commonly used adjustment factors in ADE derivations are those used to account for human

147

variability (interindividual or intraspecies variability - AFH) for the toxic response and those used for

148

interspecies extrapolation (AFA) when applying animal toxicity data to humans. In the absence of data, a

149

default adjustment factor of 10 is commonly used to account for human variability. Likewise, in the

150

absence of data, a default adjustment factor of either 10, or allometric scaling factors , are used to

151

extrapolate from an experimental animal study to humans. These consensus default factors of 10 evolved

152

from the earliest risk assessments performed by the Food and Drug Administration (FDA) where

153

biological data were limited and a factor of 10 for both interspecies extrapolation and human

154

(intraspecies) variability were combined, culminating in a “100-fold safety factor” (Lehman and Fitzhugh,

155

1954). Over the decades, as the scientific knowledge base grew and evolved, the paradigm shifted from

156

application of these default factors or allometric scaling factors to application of quantitative chemical-

157

specific adjustments, which provide a more robust scientific rationale for the overall risk assessment

158

(IPCS, 2005; Renwick, 1993, 1991; Sussman et al., 2016, this issue).

2

AC C

EP

TE D

146

159 2

Allometric scaling factors account for changes in basal metabolic rate that occur with changes in scaling animal size.

7

ACCEPTED MANUSCRIPT

The shift away from using default factors began with the proposal of Renwick (1991) to subdivide the AFH

161

and AFA default values of 10 (each) into two subfactors, one that accounts for differences in TK and the

162

other accounting for differences in TD. This subdivision serves a useful function when data are available

163

because the TK and TD data can be separately utilized to replace portions of the default value, as

164

appropriate. In a subsequent publication, Renwick (1993) determined that TK differences are typically

165

greater than TD differences and, therefore, the AFH and AFA safety factors should be unequally divided

166

into factors of 4 for TK and 2.5 for TD. The International Programme on Chemical Safety (IPCS) adopted

167

the principles set forward by Renwick (1993) but suggested that only the AFA (interspecies) factor be

168

divided unequally into 4-fold (TK) and 2.5-fold (TD), while the AFH (intraspecies) factor should be split

169

evenly (3.16 for both TK and TD) because the database analyzed was insufficient to justify an uneven

170

subdivision of the 10-fold factor for human variability (Figure 1) (IPCS, 2005).

M AN U

SC

RI PT

160

171

Based on this concept, the IPCS produced guidelines on the data needed to replace defaults with

173

empirically determined data. These chemical-specific adjustment factors (CSAF) quantify inter- and

174

intraspecies differences and variability associated with TK and TD parameters. More recently, the US

175

Environmental Protection Agency (US EPA, 2014) also introduced data-derived extrapolation factors

176

(DDEF) for intraspecies and interspecies extrapolation. The DDEF approach also moves away from the

177

10-fold framework of individual default factors (Figure 1). There are numerous papers illustrating how to

178

use and apply TK/TD data to replace default values for pharmaceuticals (Gould et al., 2013; Naumann et

179

al., 2004, 2001, 1997; Sargent et al., 2002; Silverman et al., 1999), and the guidance documents for

180

CSAF and DDEF derivation provide useful examples for the application of such data. Since the focus of

181

this communication relates to pharmaceuticals, and since the DDEF guidelines are a recent development

182

for application to environmental chemicals, the emphasis herein is on the CSAF guidelines.

EP

AC C

183

TE D

172

8

SC

RI PT

ACCEPTED MANUSCRIPT

184

Default adjustment factors, by definition, should only be used in the absence of relevant suitable data.

186

When sufficient chemical-specific data are available, CSAFs should be used to replace portions of the

187

default factors for AFA and AFH. TK and TD data are used to derive CSAFs in accordance with the

188

published guidelines (IPCS, 2005; Meek et al., 2002; Sussman et al., 2016, this issue; US EPA, 2014).

M AN U

185

189

3. CSAF Values for Intra- and Interspecies Adjustment

191

Developing CSAF values using TK data requires knowledge of the relationship between the external dose

192

and the internal target concentrations at the site of action for the critical effect. This knowledge applies to

193

the AFA differences between humans and animals and the AFH variability resulting from susceptible

194

human subpopulations. The ideal internal dose (internal exposure) would be the free drug or

195

toxicologically active metabolite concentration at the critical site of action in both humans and animals,

196

although such complete datasets are not typically available. Instead, drug concentrations in the plasma,

197

or other appropriate fluids, from either nonclinical or clinical studies, are commonly used as surrogates for

198

estimating the drug exposure at the critical site of action.

199

AC C

EP

TE D

190

200

3.1.

Choosing the Appropriate CSAF Parameter

201

The most commonly used internal dose metric is the area under the (concentration-time) curve (AUC) in

202

the fluid sampled (Figure 2). Especially for toxicants that bind covalently or cause irreversible damage, an

203

integrated measure of dose over time such as AUC is considered to be the most appropriate (US EPA,

204

2014). This parameter implicitly assumes that the effect of the chemical on the target tissue is linear over

9

ACCEPTED MANUSCRIPT

both concentration and time (Clewell et al., 2002). Less commonly, adjustments are based on an effect

206

for which peak plasma concentration is critical (e.g., interference with ion channels that can result in

207

arrhythmias or central nervous system effects), in which case maximum observed plasma concentration

208

(Cmax) is a better choice for dose metric. Other TK metrics used for CSAF determination include clearance

209

rates, glomerular filtration rates, and others that, while not direct measures of exposure, are correlated

210

with the critical target tissue concentration. Selection of the most appropriate dose metric is not always

211

easy, as it requires knowledge of the relationship between the TK profile and occurrence of the critical

212

effect. Guidance for selection of the most appropriate TK parameter and dose metric can be found in the

213

CSAF and DDEF guidance documents (IPCS, 2005; US EPA, 2014).

214

AC C

EP

TE D

M AN U

SC

RI PT

205

215

Indications that Cmax may be a good dose metric include adverse effects that are reported shortly after

216

dosing, or those that are transient and reversible, such as headache, nausea, and acute phase reactions

217

after parenteral administration. Similarly, effects such as irritancy and asphyxia are more likely to be

218

related to Cmax rather than AUC. In such instances, the concentration of chemical at the target site

219

exceeds a biological threshold concentration that acutely elicits the adverse effect (Clewell et al., 2002).

220

This could be attributable to interactions with essential cell or tissue targets that result in acute disruption

221

of biological activity, including binding with receptors (e.g., opioid receptor), enzymes (e.g., glutamine

10

ACCEPTED MANUSCRIPT

synthetase), critical cellular function proteins (e.g., mitochondrial electron transport proteins), or ion

223

channel disruption (e.g., sodium/potassium ion channels). At chemical concentrations below the critical

224

threshold, the stoichiometric interaction of the chemical with the target is insufficient to elicit a response

225

(e.g., depolarization); while at sufficiently high concentrations a sufficient proportion of the target

226

molecules (e.g., ion transport channels) are perturbed such that a response is initiated leading to the

227

observed adverse effect (e.g., seizure).

RI PT

222

228

The peak observed concentration, Cmax, is a function of the rate of absorption, volume of distribution (Vd),

230

and the rate of elimination. In the simplest theoretical circumstance, when a substance is given by rapid

231

bolus injection, Cmax can be described as the quotient of dose/volume of distribution. In reality, following

232

exposure to a substance, two processes act to lower the Cmax: elimination (metabolic, renal, etc.) which

233

removes the substance, and distribution which dilutes the substance by partitioning it to various distal

234

compartments and by binding to plasma or other proteins. For this reason, the magnitude of Cmax

235

depends on the rate at which the substance reaches systemic circulation (e.g., via absorption or infusion).

236

The time period required for a substance to reach Cmax is defined as Tmax (Figure 2). The importance of

237

the relationship between Cmax and Tmax is that a longer Tmax will typically result in a lower Cmax.

238

Conversely, a shorter Tmax can equate to a higher Cmax because there is less time for the substance to be

239

distributed and eliminated. This is particularly relevant to gavage studies where a substance is given as a

240

single oral bolus dose which results in a much shorter Tmax and much higher Cmax than if the same dose of

241

the substance were dispensed over time, such as in a dietary feeding study. For this reason, both Cmax

242

and Tmax must be considered when there is concern for a peak-related effect, and should be evaluated in

243

the context of ceiling limits as specified by the Threshold Limit Value guidelines for occupational

244

exposures established by the American Conference of Governmental Industrial Hygienists (ACGIH,

245

2016).

246

AC C

EP

TE D

M AN U

SC

229

247

3.2

Intraspecies Toxicokinetic Variability (AFH)

248

As described earlier, in the absence of data a default factor of 10 has been applied historically for AFH

249

[alternatively referred to as the F2 factor in the European Medicines Agency (EMA) guidance documents]

11

ACCEPTED MANUSCRIPT

based on a scientific rationale documented in a number of reviews (Dourson and Stara, 1983; Lehman

251

and Fitzhugh, 1954). This adjustment factor accounts for both TK and TD variations within the human

252

population (interindividual variability) and is intended to protect subpopulations that may be more

253

susceptible to toxic effects from chemical exposure due to their age, sex, genetics, pre-existing diseases,

254

etc. (IPCS, 2005). However, empirical data suggest that in some cases the application of a default factor

255

of 10 is greater than needed and thus overly stringent. When overly protective assumptions are applied

256

throughout the risk assessment process for pharmaceutical manufacturing, the cumulative consequences

257

can lead to unnecessary production costs, increased efforts related to safe handling by workers, and

258

unnecessary use of specialized production facilities. Conversely, the default 10-fold factor may not be

259

sufficiently protective. This possibility exists when known genetic polymorphisms in metabolic pathways

260

(poor metabolizers) result in higher plasma levels of the substance in those individuals than the general

261

population without the polymorphism (extensive metabolizers). Naumann et al. (2004) used timolol

262

maleate to illustrate a situation where a default factor of 10 is insufficiently protective. In this example, a

263

polymorphism in CYP2D6 results in a bimodal distribution of the human population as either poor or

264

extensive metabolizers, yielding an AFH of 9.8 for timolol maleate. When combined with the TD subfactor,

265

the resulting CSAF is 12-31 for interindividual variability, depending on the whether the default TD

266

subfactor of 3.2 is used, or a TD subfactor of 1.2 based on beta-adrenergic receptor binding data.

SC

M AN U

TE D

267

RI PT

250

The extent to which the study population is representative of the general population relies on the ratio of

269

the tail of the distribution to the central tendency for the appropriate TK parameter (IPCS, 2005; Meek et

270

al., 2002; US EPA, 2014) (Equation 1). This approach is based on the premise that if a subpopulation

271

(e.g., tail of the population distribution) is sufficiently different from the general population (i.e., a

272

significantly higher AUC or Cmax for the same dose), the acceptable exposure should to be reduced

273

(adjusted downward) to coincide with the exposure in normal (average) healthy individuals (Sussman et

274

al., 2016, this issue). Where two or more distinct subpopulations exist (bimodal distribution), the ratio of

275

the upper tail of the most sensitive subpopulation over the central tendency of the remainder of the

276

population is used to derive an appropriate adjustment factor (Naumann et al., 1997; IPCS, 2005; US

277

EPA, 2014). In either case the subfactor replaces the default AFH of 3.16 for the TK fraction, after an

AC C

EP

268

12

ACCEPTED MANUSCRIPT

278

evaluation of the sensitivity of the study population to be sure it covers the full variability for the target

279

population.    =

280

   

   

~



Equation 1.



282

3.3

283

The AFA accounts for TK and TD variations across different species and takes into account differences in

284

metabolic rates and physiological parameters between species for extrapolation. Default values for AFA

285

are variable: some guidelines recommend a factor of 10 be used, which is subdivided into factors of 4.0

286

for TK and 2.5 for TD (e.g., IPCS, 2005) (Figure 1). Other guidelines (e.g., ICH Q3C) recommend an AFA

287

adjustment factor based on allometric scaling and refer to it as the “F1” factor (ICH, 2011).

M AN U

SC

Interspecies Toxicokinetic Extrapolation (AFA)

RI PT

281

288

Allometric scaling describes the relationship between body weight or surface area and metabolic or

290

physiological differences (e.g., breathing rates, blood flows, etc.) between species. Scaling can be

291

performed based on comparative body surface area (W0.67) or on basal metabolic rate (W 0.75) (EFSA,

292

2012; ICH, 2011; Rennen et al., 2001; US EPA, 2011; US FDA, 2005). Selection of a scaling approach

293

largely depends on the regulatory agency guidelines being followed. The proposed update to the Risk-

294

MaPP guidelines (ISPE, 2010) does not specify a preference between allometric scaling based on W

295

an approach in widespread use for determining dose levels for first-in-human clinical trials (US FDA,

296

2005), or by W

297

area (W

298

basal metabolic rate approach (W

299

metabolic rate and the 0.75 exponent provides better scaling and is the preferred relationship in

300

veterinary medicine. However, the motivation for using the 0.67 exponent for human clinical trials is

301

related to safety; it provides a lower dose estimate and therefore a larger safety margin. In the absence of

302

satisfactory toxicokinetic studies, the default approach that scales an animal dose to an HED can be

303

based on body surface area, as shown in Equation 2.

,

as recommended by the US EPA (2011). The ICH Q3C guidelines specify the surface

AC C

) approach, whereas the EMA guidelines for first-in-human clinical trials refer to the US FDA 0.75

). Scientifically, body surface area is an imperfect estimate of

304 305

0.67

EP

0.67

0.75

TE D

289

The default allometric scaling values based on body surface area are described mathematically as:

13

ACCEPTED MANUSCRIPT

306

HED =  !"# $ %&'( "! )#*/,*-. × 0

1 234 )3- 78.:;. 451 234 )3-

6

Equation 2.

Allometric scaling provides a default factor that accounts only for TK differences in body surface area or

308

metabolic rate. In some cases, it is not appropriate to scale doses on the basis of body mass alone, since

309

other TK factors override the effects of scaling. Highly protein-bound drugs should be adjusted for the

310

unbound fraction before scaling; blood volume and lung surface area increase in direct proportion to body

311

mass and scale with an exponent close to 1. Chemicals that undergo active transport and those

312

undergoing significant biliary excretion or renal secretion are not likely amenable to simple allometric

313

scaling (Sharma and McNeill, 2009).

SC

RI PT

307

314

A consequence of the fact allometric adjustments capture only physiologic factors that scale with

316

metabolism (e.g., uptake and non-metabolic clearance processes) means that it may not account for

317

species-specific differences in TD (e.g., differences in target expression, receptor binding, pathway

318

activities). Allometric scaling assumes that all species are equally sensitive to the adverse effects of a

319

compound when adjusted for the metabolic rate (or body surface area) of the organisms; however, this

320

may not be the case. For this reason, there are instances where both metabolic rate and toxicodynamic

321

differences contribute to differences in sensitivity between species (Rhomberg and Lewandowski, 2004).

322

For example, if the PoD was based on toxicity studies conducted in mice and it is shown that humans are

323

substantially more sensitive to the toxicodynamic effect at the same internal exposure, it would be most

324

appropriate to apply a CSAF to account for both TK and TD factors.

TE D

EP

325

M AN U

315

When sufficient chemical-specific data are available, deriving a CSAF for AFA provides a more robust and

327

scientific approach for deriving non-default values than allometric scaling factors (IPCS, 2005). The

328

assumption is that humans are more sensitive than the animal species from which the PoD was derived,

329

and by employing an adjustment factor to address AFA differences the ADE is reduced accordingly. In

330

some cases, animals may be more sensitive, as is the case when higher enzyme activity results in

331

greater bioactivation of a less toxic parent substance to more toxic metabolites.

AC C

326

332 333

As with the AFH, an initial decision is made with regard to whether the critical effect is likely to be related

14

ACCEPTED MANUSCRIPT

to the Cmax or to the AUC. In general, it is reasonable to assume that effects that emerge over time (i.e.,

335

with subchronic or chronic exposure) are related to the AUC, whereas acute effects could be related to

336

either AUC or Cmax. The adequacy of the available data is considered by taking into account aspects of

337

the critical studies such as the routes of administration, nature of the population, administered doses, and

338

sample size. The numbers of animals and humans in the dataset should be sufficient to ensure that the

339

data allow a reliable estimate of the central tendency for each species. As a guide, the standard error

340

[standard deviation (SD) of the sample divided by the square root of the sample size] should be less than

341

approximately 20% of the mean. The CSAF is calculated by the ratio of the kinetic parameter in animals

342

divided by the same kinetic parameter in humans (Equation 3), assuming the chosen parameter is

343

properly adjusted for confounding factors such as dose, administration frequency, period of integration

344

(length of time used in calculating the AUC), etc. In some instances, the toxicokinetics dataset may not

345

allow satisfactory evaluation of Cmax and AUC parameters, but does establish clearance rate. In such

346

cases, clearance can also be used to calculate a CSAF; however, it is important to note that clearance

347

rates are inversely related to AUC and Cmax [clearance = (bioavailability x dose)/AUC], therefore, the

348

greater the clearance, the lower the AUC and Cmax. For this reason, the clearance ratio is the reciprocal

349

(animal / human) to AUC and Cmax, as shown in Equation 3.

351

SC

M AN U

TE D

350

RI PT

334

<   =

=>? @ F @
×

@ HIJK

@ JKLIJM

Equation 3.

4. Bioavailability and Route Specific Adjustments Using TK/TD Data

353

The relative bioavailability of a substance via the administration route in the critical study and the route of

354

interest can be used to adjust the PoD to reflect differences in route-specific bioavailability. Bioavailability

355

is a measure of internal exposure that describes the fraction of a chemical or its active moiety that is

356

systemically absorbed and distributed in the body. Route-to-route extrapolation is necessary when

357

attempting to derive an ADE from a study conducted by one route (e.g., oral) that is different from the

358

potential route of exposure (e.g., intravenous). Although direct studies using the relevant route of

359

administration are preferred, extrapolation using a bioavailability correction factor may be useful to ensure

360

adequate protection in these situations (Naumann et al., 2009). To illustrate the importance of

361

bioavailability corrections, consider the situation where an ADE for product A (an oral drug) was used to

AC C

EP

352

15

ACCEPTED MANUSCRIPT

support cleaning limits for product B (an intravenous drug) in an API manufacturing facility. The oral

363

bioavailability of product A is 1%, i.e., only 1% of the administered dose is systemically available in the

364

blood. If product B is administered parenterally but this difference in route is not accounted for, the

365

amount of allowable carryover would be two orders of magnitude higher for the parenteral dosage form

366

(given that product A will have 100% bioavailability from the parenteral route). For this reason, the ADE

367

for product A that is protective for parenteral exposure is 100-fold lower than the ADE for the oral route.

RI PT

362

368

Route-to-route extrapolation is most relevant for relating non-local effects to systemic exposure levels

370

(Sargent et al., 2013). To relate external dose to internal dose, a correction factor is used to account for

371

differences in bioavailability by different routes, particularly for pharmaceutical industry OELs where the

372

critical effect is typically derived from oral or parenteral studies and the primary exposure of concern is

373

inhalation. Assessing the ratio of the same toxicity endpoint via multiple routes of administration for the

374

same chemical can also assist in establishing whether toxicity is greater via one route versus another. For

375

example, if the NOAEL for a study conducted via the oral route is 30 mg/kg-day, and the NOAEL is 3

376

mg/kg-day for a similar study conducted via the intravenous route, decreased bioavailability via the oral

377

route may account for this 10-fold difference. In the absence of data, a protective default assumption of

378

100% bioavailability is often assumed for the inhalation, subcutaneous, dermal, and all parenteral

379

exposure routes for small molecules, unless there is evidence indicating otherwise. There are a number

380

of route-specific metabolic considerations related to bioavailability, as discussed in the following sections.

EP

TE D

M AN U

SC

369

381 4.1

Default Adjustments for Individual Route of Exposure

383

4.1.1

384

Lower respiratory tract deposition is considered to be more relevant to overall bioavailability of inhaled

385

chemical aerosols than the total inhaled dose, and intra-nasal deposition or instillation often (but not

386

always) has limited bioavailability. Including data on the particle size distribution, potential regional

387

distribution from inhalation, and solubility in PBPK modeling could greatly increase the accuracy of OELs

388

and ADEs for inhalation exposure over the highly precautionary practice of assuming 100%, which is

389

currently practiced by many organizations (Naumann et al., 2009; RIVM, 2002). Dosimetry models

AC C

382

Inhalation

16

ACCEPTED MANUSCRIPT

390

available to assist in estimating deposition of aerosols are available for risk assessment (Kuempel et al.,

391

2015). Although dosimetry methods such as computation fluid dynamics are available for assessing gas

392

phase exposures, such exposures are less common for pharmaceuticals.

RI PT

393 Inhalation differs from other routes of exposure because determining the portion of a dose delivered to

395

the deep lungs, where most absorption occurs, is a complicated matter. Depending on the size and

396

aerodynamics of the inhaled particles, inhaled aerosols can deposit in the nose, mouth, throat, upper and

397

lower airways, and deep lung, and a fraction of the inhaled mass typically escapes during exhalation.

398

Particles >10 µm can be expected to deposit via sedimentation or impaction in the nasopharynx and

399

tracheobronchial passages. In humans, particles that deposit in the upper airway are generally cleared

400

via the mucocilliary escalator and subsequently swallowed, which can, for some molecules, provide a

401

secondary (oral) route of exposure. Particles with sizes in the ranges of 1–5 µm in diameter are most

402

efficiently deposited in the deep lungs (pulmonary region), with >50% of the 3 µm diameter particles being

403

deposited in the alveolar region (Labiris and Dolovich, 2003). With the growing interest in biologic

404

pharmaceuticals, occupational exposure to inhaled polypeptides and proteins has become a growing

405

concern. For large therapeutic proteins (> 40 kD), there is evidence that the bioavailability by inhalation is

406

maximally 5% (Pfister et al., 2014), while the oral bioavailability is negligible in the gastrointestinal tract

407

due to proteolysis.

409

4.1.2

410

The gastrointestinal (GI) tract subjugates substances to a combination of conditions not associated with

411

any other route of administration. GI flora and enzymes released into the GI tract mediate chemical

412

changes including hydrolysis, oxidation/reduction, and conjugation; lipophilic substances are emulsified

413

for increased intestinal absorption; a host of transport systems are present that efficiently move

414

substances across membrane barriers; and well perfused microvilli dramatically increase the surface area

415

available for absorption. In addition, local conditions in various regions of the GI tract can alter the

416

ionization state of substances (e.g., weak acids and bases), which can facilitate absorption in the

417

stomach where pH levels are below the pKa values of many weak acids (ECHA, 2010).

AC C

Oral

EP

408

TE D

M AN U

SC

394

17

ACCEPTED MANUSCRIPT

In the absence of specific data on oral bioavailability, defaults are often suggested. ECHA (2010) has

419

recommended a bioavailability default of 50% in the absence of data when there is evidence that there is

420

no first-pass effect, reabsorption, or bolus effect (Sargent et al., 2013). Surrogate data and potential ways

421

to predict oral absorption and bioavailability are still proving difficult to model, with few, if any, simple rules

422

showing promise in offering a qualitative way to set defaults. Tian et al. (2011) showed that rule-based

423

systems [e.g., Lipinski’s rule of five; Lipinski et al., (2001)] used for predicting absorption based on

424

molecular properties [e.g., molecular weight, partition coefficient (Log P), water-solubility, etc.] are not

425

particularly good predictors of oral bioavailability because they do not account for the influence of

426

important metabolic processes, such as liver metabolism. Rather, such simple rules only show good

427

performance for classifying intestinal absorption, not for classifying oral bioavailability. In fact, molecules

428

with low metabolism are more closely correlated with model predictions than those with higher metabolic

429

reactivity. This potentially underscores the importance of first-pass metabolism in any model that hopes to

430

predict bioavailability via the oral route (Tian et al., 2011).

M AN U

SC

RI PT

418

431

First-pass metabolism is a form of pre-systemic clearance that can be a critical determinant of systemic

433

bioavailability for parent molecules. First-pass metabolism decreases the apparent absorption of a

434

chemical before the chemical reaches the systemic circulation. In some cases, however, first-pass

435

metabolism can rapidly transform a less toxic parent molecule into the active toxicant (metabolite)

436

responsible for adverse effects in tissues distant from the site of metabolism (e.g., cyclophosphamide). In

437

theory, first-pass metabolism can occur via any route of administration where enzyme systems are

438

present in sufficient quantity to prevent absorbed substances from reaching systemic distribution,

439

including the lungs, skin, and other tissues. However, the liver is the primary site for first-pass metabolism

440

due to its importance as a site of biotransformation and as the portal of entry for substances carried by

441

the mesenteric blood flow from the GI tract. Pulmonary metabolism of some substances also occurs

442

(Borlak et al., 2005), but few substances are reported to undergo a quantitatively important pulmonary

443

first-pass metabolism (ECHA, 2010).

AC C

EP

TE D

432

444

18

ACCEPTED MANUSCRIPT

First-pass metabolism is a major consideration impacting bioavailability from the oral [and intraperitoneal

446

(IP)] routes of exposure that is not typically associated with other routes of exposure. Liver first-pass

447

metabolism can dramatically reduce systemic exposure to substances (or increase exposure to

448

metabolites) by the oral and IP routes of administration. When a substance is administered by either of

449

these routes, it is absorbed primarily via the mesenteric circulation, which, after perfusing the small

450

intestines and a portion of the colon, empties to the portal vein and is delivered to the liver. In this way,

451

100% of the mesenteric blood flow, and all substances absorbed from the intestines or peritoneal space

452

pass through the liver before entering the general circulation (as parent compound or metabolites). As a

453

result, the same dose administered via another route (e.g., inhalation, intravenous) will result in

454

substantially higher systemic exposures, as measured by Cmax and AUC. In general, oral studies do not

455

provide sufficient data to distinguish the relative contributions of absorption vs. first-pass metabolism to

456

decreased bioavailability, and both are observed jointly as a relative decrease in systemic exposure. It is

457

also important to recognize that the liver can be a target of toxicity in the absence of systemic exposure.

458

Since the liver receives 100% of the mesenteric blood flow, a chemical substance can be absorbed from

459

the GI and delivered to the liver where it exerts its toxicological effect. This can be an effect of the parent

460

chemical or as a result of an active metabolite. Administration of the same chemical via other routes could

461

likely produce a similar effect; however, systemic delivery to the liver would be required, albeit at lower

462

plasma concentrations due to dilution, and in such cases, other target tissue effects might also be

463

observed.

SC

M AN U

TE D

EP

464

RI PT

445

Liver metabolism may be either a low extraction (capacity-limited) process, in which the enzymatic

466

capacity of the liver is the limiting factor, or a high extraction (blood flow-limited) process, where the

467

limiting factor for the rate of metabolism is the rate at which the chemical is delivered to the liver. The

468

expression and activity of many hepatic metabolizing enzymes can be perturbed by a host of factors

469

including age, environmental factors (e.g., diet, lifestyle), gender, pregnancy, and concurrent

470

pharmaceutical use. When enzyme systems mediating first-pass metabolism are perturbed by any such

471

factors, the effect is usually in the direction of a reduction in metabolizing capacity for high extraction

472

chemicals since the responsible metabolic pathways are already performing with high efficiency;

AC C

465

19

ACCEPTED MANUSCRIPT

therefore, perturbations that increase activity have little additional effect on clearance while inhibition can

474

dramatically reduce efficiency. Conversely, perturbations of the activity of metabolizing enzymes (e.g.,

475

enzyme induction) have the greatest impact on low extraction chemicals that undergo capacity-limited

476

clearance since these perturbations can both increase or decrease the metabolizing activity of these

477

systems (Fan et al., 2015). Once a chemical is systemically available, the kinetic disposition is the same

478

regardless of exposure route.

RI PT

473

479

It should be noted that chemicals with a PoD that is derived locally at the initial site of contact will have

481

route-specific toxicity. In these cases, route-to-route extrapolation is not considered appropriate. For

482

example, when irritancy is the concern, the effect is often dependent on concentration at the site of

483

contact rather than systemic dose. When route-specific toxicity exists, it may be necessary to establish

484

both a concentration limit for the chemical in question as well as a systemic ADE. Another situation in

485

which route-to-route extrapolation is not considered appropriate is hypersensitivity, where the types of

486

reactions tend to be route specific (Gould et al., 2016, this issue).

M AN U

SC

480

487 4.1.3

489

There are a number of parenteral routes of administration including: intravenous, intramuscular,

490

subcutaneous, intraosseous, intraventricular, epidural, intracardiac, intraarticular, intracavernous,

491

intravitreal, and others. Only a few of these are common administration routes, but similar considerations

492

on adjusting for route of exposure should be applied to each. In many cases, standards exist for daily

493

intake of substances in food, water, air, and occupational exposure but without information for parenteral

494

administration. Where data are available but not considered sufficient for a safety assessment, the ICH

495

Q3D guideline for elemental impurities in pharmaceutical products lays out the following guidelines for

496

using oral standards for parental ADEs (ICH, 2013):

AC C

EP

Parenteral Routes

TE D

488

497



498



Oral bioavailability < 50% - divide by a modifying factor of 10

499



Oral bioavailability between 50% and 90% - divide by a modifying factor of 2

500



Oral bioavailability > 90% - divide by a modifying factor of 1

Oral bioavailability < 1% - divide by a modifying factor of 100

20

ACCEPTED MANUSCRIPT

501

However, there are some limitations associated with these default values for general application. The

502

guidelines do not address what to do when there are no data indicating absolute bioavailability via the

503

oral route, or how potential oral bioavailability should be estimated.

505 506

4.2

RI PT

504 Bioavailability Correction Factor (BCF) to Replace Default Adjustments for Route of Exposure

Absolute bioavailability (F) is defined as the dose-normalized bioavailable fraction by any extravascular

508

route (e.g., oral, dermal, etc.) divided by the dose-normalized intravenous bioavailable fraction (presumed

509

to be 100%) (Equation 4).  = 100 ×


×

@ LS

@ PQHR

Equation 4.

M AN U

510

SC

507

Relative to direct intravenous administration, other routes can have some degree of absorption

512

inefficiency resulting in either decreased or delayed systemic bioavailability. In some instances, when

513

data from the relevant route are not available, bioavailability adjustments are useful to ensure adequate

514

exposure protection. If the differences are quantitatively relevant, a bioavailability correction factor (BCF)

515

should be applied (Naumann et al., 2009), resulting in route-specific ADE values. A BCF, also referred to

516

as alpha (α) in some references (e.g., Sargent and Kirk, 1988), is defined as the bioavailability via the

517

exposure route of interest divided by the bioavailability via the route used in the critical study (Naumann

518

et al., 2009), as shown in Equation 5. The BCF is equivalent to 1/F and is included in the denominator of

519

the limit derivation.

EP

520

TE D

511

TU &V $Wℎ Y. =

% [@\] ^ \ 4 E@ 5 @5  .3.,4 @. % [@\] ^ \ 4    5`^ @5  .3.,@.

Equation 5.

In practice, the ADE is derived from the PoD based on the route used in the critical study by placing the

522

BCF (α) in the denominator of the typical ADE equation adapted from Naumann et al. (2009). As an

523

example, consider an ADE derived based on an effect observed from an oral route of administration. If

524

the systemic bioavailability for the oral route is 10% and the bioavailability by the inhalation route is

525

assumed to be 100%; the resulting BCF would be 100/10 or a factor of 10. Thus, application of this factor

526

lowers the ADE by a factor of 10 to account for the greater bioavailability via the inhalation route. This

527

adjustment estimates the intravenous equivalent of the oral PoD on an internal dose basis, and would be

528

relevant to a manufacturing scenario where a manufacturing line was used to produce an oral drug which

AC C

521

21

ACCEPTED MANUSCRIPT

529

was followed by production of a parenteral drug. Likewise, a similar situation would occur if a PoD is

530

based on oral dosing studies but the concern is for occupational inhalation exposures.

531

533

There are a number of special considerations to be taken into account when applying a BCF:

RI PT

532

1. When extrapolating from oral or intraperitoneal animal data to other routes (e.g., inhalation,

534

dermal) between species, consideration should be given to differences in the rate and extent of

535

liver metabolism (e.g., first-pass clearance) between humans and the study species.

2. The rate of absorption can be very different for different routes of exposure. For example, in

537

humans, oral and dermal exposure tends to be slower than inhalation. For this reason,

538

consideration should be given to effects from inhalation associated with peak plasma

539

concentrations (Cmax), since quicker absorption increases peak systemic concentrations despite

540

comparable AUC values.

M AN U

541

SC

536

3. Rates of absorption can be limited by saturation due to limited capacity of enzymes and transporters involved in the biotransformation or absorption processes. As a result, the PoD may

543

be based on a non-linear dose-response relationship (ECHA, 2010).

544

TE D

542

Initially, an ADE should only be calculated for the route with the best available data. This initial ADE

546

should be the starting point for calculating ADEs for alternative routes of exposure, if such ADEs are

547

needed. As route-specific bioavailability may also differ between species, ADE calculations for these

548

alternative routes of exposure should be made based on comparative human exposure data (e.g., results

549

from an absolute bioavailability study), wherever possible. There have been a number of compilations for

550

drugs of both animal and human oral bioavailabilities which may prove useful (Akabane et al., 2010; Cao

551

et al., 2006; Chiou and Buehler, 2002; Musther et al., 2014; Sietsema, 1989). If experimental human data

552

are missing, route-specific differences in bioavailability may be estimated using read across from

553

chemicals with closely related structure, physiochemical properties, and toxicity profiles.

AC C

EP

545

554 555

Several investigations and reviews have provided background on the use of BCFs and guidance on when

556

and how they should be applied for OEL development (Naumann et al., 2009; Pfister et al., 2014; Sargent

22

ACCEPTED MANUSCRIPT

et al., 2013). These same approaches are relevant to setting ADEs. In many cases, the absolute oral

558

bioavailability of a compound may not be known, as it may not have been administered to humans by the

559

intravenous route. However, it is always preferable to use whatever reliable data are available for a

560

compound, rather than apply a default correction factor.

RI PT

557

561

Interspecies differences in oral bioavailability are most commonly a result of differences in first pass

563

metabolism in the gut and liver (Clark and Smith, 1984; Musther et al., 2014). Although it is well

564

established that the metabolic enzymes responsible for the biotransformation of drugs can be very

565

different between species, it does not necessarily mean that oral bioavailability will always be very

566

different. The percentage of drugs for which the human and animal oral bioavailabilities differed by more

567

than 10-fold for the same drug were lowest for dogs at 2% and rats/rodents at 2%/8%, while monkeys

568

demonstrated the greatest differences at 11%, 38%, and 40%, as measured by three different groups

569

(Akabane et al., 2010; Chiou and Buehler, 2002; Sietsema, 1989). The large difference between human

570

and primate bioavailability may, however, reflect the much more limited datasets (N = 35, 13, and 10,

571

respectively) available for primates compared with the datasets available for other species. A recent

572

reanalysis using the largest bioavailability dataset to date, inclusive of most of the individual datasets

573

indicated above, confirmed the poor overall correlation between human and animal oral bioavailability

574

(Musther et al., 2014), strongly supporting application of BCFs when data are available. In this way, the

575

BCF adjustment reduces uncertainty by increasing or decreasing the PoD as dictated by the available

576

data. It is noteworthy that the predictive value of animal bioavailability data is improved for substances

577

that have good membrane permeability, are not drug transporter substrates, and undergo little or no in

578

vivo metabolism (Akabane et al., 2010).

M AN U

TE D

EP

AC C

579

SC

562

580

5. Time Weighted Averaging, Study Duration, and Bioaccumulation Adjustments

581

Frequently, the critical study used to identify the PoD and derive the ADE has either an infrequent dosing

582

schedule or is from a subchronic duration study. As the ADE is to be protective of daily lifetime exposure,

583

the PoD derived from a study with infrequent dosing (e.g., once weekly administration) should be

584

adjusted for everyday exposures. In the absence of data, Haber’s Rule (see below) has been used by

23

ACCEPTED MANUSCRIPT

convention. However, when data are available, the preferred approach is to use TK data to assess steady

586

state and bioaccumulation through derivation of a steady state “S” factor (Sargent and Kirk, 1988).

587

Likewise, when the critical study used is not from a chronic study, a subchronic to chronic adjustment

588

factor (AFS) is typically applied on the assumption that the adverse effects of chemical exposure will occur

589

at lower exposure dose levels or with greater severity as the exposure duration increases. This can be

590

due to both the accumulation of the compound and/or the damage that it produces. While the AFS cannot

591

always be replaced by application of an S factor, it may be appropriate to use a reduced default AFS

592

pertaining to study duration (subchronic to chronic) when an S factor is used. These factors are related

593

and not independent, as the S factor differs from the AFS in that it only addresses TK considerations.

SC

RI PT

585

M AN U

594 5.1

596

Haber’s rule (Fritz Haber, 1868 - 1964) states that the concentration (C) of a substance multiplied by the

597

length of time (t) it is administered produces a fixed level of effect for a given endpoint, and thus predicts

598

the severity and incidence of the adverse health effect. Haber’s rule is a standard convention used for

599

dose averaging chemicals to make duration adjustments to the PoD when the administration frequency

600

used in the critical study is different from the exposure scenario. Haber’s rule provides an approximation

601

relating exposure to effect; however, there are many exceptions to the rule, such as when the adverse

602

effect results from Cmax or when exposure is for less than the biological half-life. Thus, understanding of

603

the chemical’s mechanism of action is an important consideration when deciding to use Haber’s rule.

EP

604

Time Weighted Averaging for Dose Frequency - Haber’s Rule

TE D

595

For intermittent exposures or exposures with variable concentrations, the average concentration of

606

exposure over the time period is used, such that concentration x time is equivalent to the AUC (Gaylor,

607

2000). For interpolation between different short-term exposures, this method may prove to be more

608

appropriately protective than when extrapolating to longer times [where t (actual exposure) / t (study

609

exposure) is greater than one]. However, in the absence of data for different durations of exposure,

610

extrapolating to shorter exposure may be inappropriate (Gaylor, 2000). A very important exception to

611

Haber’s rule is when toxicity is associated with peak concentration levels (Cmax).

AC C

605

612

24

ACCEPTED MANUSCRIPT

5.1.2

Examples

614

The PoD of 20 mg/day is from an IV infusion study in which patients received an anti-neoplastic drug 6

615

hours a day for 3 days during the first week, followed by 3 weeks without treatment (total 4 weeks or 28

616

days for the entire regimen). Developing an ADE using dose averaging, Haber’s Law states to multiply

617

the PoD by 3/28 (3 days exposure in the study/28 day exposure scenario). For example: 20

618

13

`^

×0

b `^

c `^

6 = 2.1

13

`^

de

RI PT

613

Haber’s law offers an estimated duration adjustment in the absence of data; however, the preferred

620

approach is to use TK data to assess steady state bioaccumulation, and MOA data to assess concerns

621

associated with peak effects (as described in section 3.1). In this respect, the anti-neoplastic regimen

622

described above might provide an AUC of 1500 ng•hr/mL. A similar operation to that described above is

623

used to adjust for the dosing frequency and duration. For example: 1500

M AN U

SC

619

!* ∙ ℎV 3 % k' !* ∙ ℎV ×i m = 375 de #h 28 % k' #h

In both instances either the dose or AUC could serve as the PoD. Additional adjustments for route of

625

administration (i.e., BCF) and steady state accumulation (described below in section 5.3) would be

626

applied if required.

627

TE D

624

5.2

Study Duration Adjustment Factor (AFS)

629

According to the Risk-MaPP guidance, ADEs are calculated for a potential lifetime exposure (ISPE,

630

2010). Although this is a highly protective assumption in most circumstances, the guidelines nevertheless

631

state that if the PoD is from a study that is less than lifetime, an adjustment factor may need to be applied

632

(AFS) (Dourson et al., 1996; Sussman et al., 2016, this issue). This duration adjustment for the critical

633

study is based on the assumption that the adverse effects of chemical exposure will occur at lower dose

634

levels as the exposure duration increases. For example, one could use a short-term animal study as the

635

basis for a chronic exposure ADE in humans. Considering that some drugs are often prescribed to

636

patients with intermittent dosing, such as once weekly, it is plausible that a worker or patient could be

637

exposed on a daily basis to a drug that is intended for weekly, biweekly, or monthly human dosing. But

AC C

EP

628

25

ACCEPTED MANUSCRIPT

638

the guidance documents available for these conversions are not robust enough to cover all extrapolation

639

scenarios.

640 The ICH Q3C guidelines for solvent impurities in drug products sets specific default factors for use when

642

adjusting from a shorter term study to a lifetime exposure, based on study species and the fraction of

643

animal lifetime covered in the study (ICH, 2011). No guidance document specifically addresses what data

644

are necessary to replace duration adjustment defaults or of what magnitude they should be adjusted

645

when certain TK data are available. The Risk-MaPP baseline guide states that in the absence of TK data

646

for duration adjustments, a factor of 3 is sufficient to account for the possibility that a lower PoD would

647

potentially have resulted from a longer-term study (ISPE, 2010). Risk-MAPP also suggests that when

648

longer studies are used to set shorter term limits, an upward adjustment to the ADE using an adjustment

649

factor of less than 1 may be appropriate. It does not, however, give guidance on how to calculate a

650

fractional adjustment factor or what is most appropriate (ISPE, 2010).

M AN U

SC

RI PT

641

651

Naumann and Weideman (1995) noted that high quality toxicology studies support an adjustment factor

653

of 3 to account for the possibility that a lower NOAEL could have resulted if longer-duration studies were

654

conducted (Kadry et al., 1995; Lewis and Nessel, 1994; McNamara, 1976; Weil and McCollister, 1963;

655

Woutersen et al., 1984). A recent study that tried to minimize chemical- and study-specific variability

656

identified an appropriate adjustment factor of 3 to 4 for orally administered compounds in the 90

657

percentile, with similar values for inhalation studies (Batke et al., 2011). Since these evaluations are

658

based primarily on data derived from studies where steady state was obtained, effect progression largely

659

reflects TD and TK effects other than accumulation (i.e., redistribution, latency before damage, age-

660

related changes, etc.). Hence, if an adjustment factor of 3 or 4 covers effect progression, then, in the

661

context of a traditional default factor framework where a full factor is 10x, the remaining factor for TK

662

variability is approximately 3x. A 3x factor is appropriate for accumulation for chemicals where the half-life

663

is about 2-fold greater than their administration frequency; a longer half-life or shorter administration

664

frequency will result in greater than 3-fold accumulation.

th

AC C

EP

TE D

652

665

26

ACCEPTED MANUSCRIPT

5.3

Data-Based Steady State “S” Factor to Account for Bioaccumulation and Portions of AFS

667

A bioaccumulation adjustment is required when an ADE is developed on the basis of single-dose or short-

668

duration studies and applied to a more protracted exposure scenario, and can be based on available

669

chemical-specific TK data. The steady state internal concentration can be calculated for one compartment

670

models using elimination half-lives (or from the half-life of the beta-phase of the elimination curve in two

671

compartment models) and other TK data. Steady state systemic concentrations are considered to be

672

achieved in multiple-dose studies when the period over which they are conducted exceeds 3 to 5

673

elimination half-lives (~90% to ~97% of steady state, respectively) of a chemical substance, after which

674

little additional accumulation will occur with additional administrations (as long as the dosing remains

675

consistent) (Rowland and Tozer, 1980a).

M AN U

SC

RI PT

666

676

For the development of ADEs, it can be assumed that the substance is effectively cleared if repeated

678

exposure occurs every 1.44 half-lives or less (e.g., daily exposures or work shifts) (Rowland and Tozer,

679

1980b). If the PoD is taken from a short or single-dose study and the substance has a long t½, then

680

bioaccumulation can be expected. To reduce the dose at the PoD in order to account for accumulation

681

from repeated administration, a bioaccumulation factor or steady state factor is applied (Sargent et al.,

682

2013). The S factor is a ratio of the plasma levels of the substance after steady state is achieved in a

683

repeat-dose study to the plasma levels following a single dose (ISPE, 2010). Since the S factor is used in

684

the denominator, the ADE is reduced by the incremental increase in blood levels following repeated vs.

685

single exposure. For example, a 30-day study is used as the critical study and the chemical has a 15-day

686

half-life. With daily administration, steady state would not be achieved for 2-3 months and the

687

accumulation ratio would be predicted to be approximately 21-fold. This adjustment is not needed when

688

the PoD is from a study in which steady state has been reached.

EP

AC C

689

TE D

677

690

If the t½ of the drug is known from a single-dose or short-term study, but its steady state profile is not, the

691

extent of drug accumulation can be estimated (Rowland and Tozer, 1980b) using Equation 6:

692

7.ss E t½

oop#p$ q"&! V q"& r. = @ 3  \

693 27

Equation 6.

ACCEPTED MANUSCRIPT

It is acknowledged that the above equation may have some degree of error since it is most applicable for

695

substances described by single compartment kinetics, whereas most substances are described by a TK

696

model with two or more compartments (Gibaldi and Perrier, 1982). However, for most drugs the

697

distribution phase is much shorter than the elimination phase, and so the extent of drug accumulation will

698

be mainly dependent on the elimination t½ and the calculated accumulation ratio will be a good

699

approximation (Dettli, 1982).

RI PT

694

700

While the AFS is applied when the PoD is based on a less than lifetime critical studies, it is intended to

702

generally account for TD considerations (the potential for the effects to have inadequate repair time) and

703

drug accumulation. In the presence of actual data (e.g., clearance), it is more systematic to apply an S

704

factor to account for bioaccumulation at steady state. It is important to stress that caution must be taken

705

not to overlap the AFS with the application of the S factor; otherwise accumulation would be accounted for

706

twice. However, S factor differs from the AFS default in that it the S factor only addresses TK

707

considerations, not TD-related changes. Notwithstanding, these factors are related and not independent.

708

The discrimination between the accumulation of systemic exposure up to steady state and the

709

accumulation of the related biological effect is often not possible. In practice, both aspects are taken into

710

account by applying the factor for study duration. Carefully describing the considerations involved in

711

applying each factor allows for greater transparency and will reduce the likelihood of inadvertently

712

accounting for the same factor twice.

713

EP

TE D

M AN U

SC

701

6.

Summary and Conclusions

715

The focus of this paper was how TK/TD data can be used to derive ADEs and other safe limits. It is

716

expected that this paper be read in the context of the series of papers in general and closely along with

717

the AF paper (Sussman et al., 2016, this issue), as the larger context for these adjustments is presented

718

there. Overall, ADEs and OELs are not meant to be developed by non-professionals, thus this paper is

719

not meant to be standard operating procedure for the derivation of ADEs. Also, every ADE should be

720

accompanied with full documentation of the data used, the rationale for the decisions made, and the

721

calculations at each step.

AC C

714

28

ACCEPTED MANUSCRIPT

Health-based risk assessment methodologies are now the standard of practice for setting ADEs for APIs

723

and other impurities (EMA, 2014; ICH, 2013, 2011; ISPE, 2010). Guidance documents advocate the use

724

of chemical-specific health-based approaches for setting exposure limits beginning with a rigorous critical

725

evaluation and synthesis of all pharmacology and toxicology data. Default factors have traditionally been

726

used to account for interindividual variability adjustment (AFH) and uncertainties associated with

727

extrapolation from animals to humans (AFA). In the absence of toxicokinetic data, a default adjustment

728

factor of 10 is commonly used to account for each of these such that the total adjustment factor applied to

729

the ADE is 100-fold (10 AFA x 10 AFH). Allometric scaling approaches have also been used to extrapolate

730

from animals to humans; however, such an approach accounts only for differences in how body surface

731

area or body weight scale to metabolic rate. Recall that this latter factor assumes humans are more

732

sensitive than animals, which, while a protective assumption, may not be true for all compounds. Also,

733

some factors, such as blood volume and renal secretion, do not predictably scale with body size (Sharma

734

and McNeill, 2009).

M AN U

SC

RI PT

722

735

When sufficient data are available, data-driven CSAF approaches have been described and provide

737

detailed guidance on how to use TK and TD data in place of default AFA and AFH adjustment factors

738

(IPCS, 2005; Meek et al., 2002; US EPA, 2014). The CSAF guidance subdivides the AFH factor into two

739

subfactors that separately account for toxicokinetic and toxicodynamic variability. These factors account

740

for variability in the dose metric selected as most relevant for the critical effect, and guidance on selection

741

of the appropriate dose metric is also available (IPCS, 2005; US EPA, 2014).

EP

AC C

742

TE D

736

743

Derived ADE limits can also consider adjustmenting for route-specific differences in bioavailability.

744

Although it is preferable to use studies most relevant to the expected route of exposure, such studies are

745

not always available. In these situations, it is useful to apply a BCF to extrapolate the PoD from the route

746

used in the critical study to the route of the anticipated exposure. Once a chemical is systemically

747

available, the kinetic disposition is the same regardless of route of exposure, although there are many

748

physiological factors that impact systemic availability.

749

29

ACCEPTED MANUSCRIPT

The risk assessment process also takes into consideration the duration of the critical study in which the

751

critical effect is identified, and the dosing schedule and frequency of dose administration in the critical

752

study. Haber’s Rule has been traditionally used to average infrequent dosing schedules over time.

753

However, when data are available, the steady state S factor can be used instead to account for potential

754

accumulation from a different dosing scenario. In instances where short-term and subchronic studies are

755

used as the basis of the ADE, an adjustment factor (AFS) is applied to account for the short length of the

756

study and the possibility that a longer study might yield a lower PoD or an effect of greater severity at the

757

same dose. When the PoD is from a short-term study (i.e., acute, subchronic) or when the substance has

758

a half-life that is significantly longer than the dosing frequency, the preferred approach is to calculate the

759

accumulation ratio (S factor) to adjust the PoD for predicted bioaccumulation. In doing so, a reduced

760

adjustment factor for subchronic to chronic extrapolation (AFS) should be applied. The application of a

761

separate factor to account for steady state accumulation with longer durations of exposure has several

762

advantages:

M AN U

SC

RI PT

750

1. There is no established guidance on how to subdivide the adjustment factor for study length on

764

the basis of TD and TK components. In the absence of an accepted authoritative guideline,

765

current practices are ad hoc and inconsistent.

TE D

763

2. Accounting for accumulation with a separate bioaccumulation ratio increases transparency and

767

can easily permit the use of longer studies to set shorter term limits, and the upward adjustment

768

of the ADE in the absence of accumulation for drugs that are cleared rapidly (Sargent et al.,

769

2013).

771 772 773 774 775 776

3. Reserving the adjustment factor for subchronic to chronic extrapolation for toxicodynamic effects

AC C

770

EP

766

permits greater flexibility to consider reductions in the NOAEL/LOAEL boundary and increasing effect severity with increasing exposure duration. The average differences between subchronic and chronic values are in the range of 3-fold, whereas a small percentage of chemicals have ratios exceeding 10-fold (Naumann and Weideman, 1995).

4. Application of a separate bioaccumulation ratio helps to minimize overlap among adjustment factors and supports independence among adjustment factors.

777

30

ACCEPTED MANUSCRIPT

778

All of these factors are tied together into a single formula used for derivation of ADE limits, as shown in

779

Equation 7: x@.[y.

780

vw = 
781

where the PoD is in mg/kg; BW is in kg; AFC is the composite adjustment factor including all CSAF

782

adjustments and other adjustments); BCF is the unitless bioavailability correction factor; S is the steady-

783

state adjustment ratio; and MF is any additional required modifying factor(s) (Naumann et al., 2009). The

784

use of TK and TD data in the derivation of the ADE is the most data-based and robust approach to the

785

protection of human health.

SC

RI PT

Equation 7.

786 7. Acknowledgements

788

The statements and conclusions in this paper reflect the opinions of the authors and do not necessarily

789

represent official policies of the organizations as listed on the title page. The authors would like to

790

acknowledge Patricia Weideman, Andrew Maier, and Alison Pecquet for organizing and facilitating the

791

workshop that served as the basis for developing this manuscript. The authors would also like to thank all

792

of the participants of the workshop for their contributions at the workshop and subsequent reviews of this

793

manuscript, including: Joel Bercu, Courtney Callis, David Dolan, Andreas Flueckiger, Janet Gould, Eileen

794

Hayes, Robert Jolly, Ester Lovsin Barle, Wendy Luo, Eric Morinello, Lance Molnar, Michael Olson,

795

Christopher Seaman, Claudia Sehner, Bryan Shipp, Brad Stanard, Robert Sussman, and Andrew Walsh.

796

The manuscript was developed in part with funding from Genentech Inc. for organizational and editorial

797

staff activities.

798

8. References

799

ACGIH, 2016. TLV/BEI Development Process. American Conference of Governmental Industrial

801

TE D

EP

AC C

800

M AN U

787

Hygienists. Available at: http://www.acgih.org/tlv-bei-guidelines/policies-procedurespresentations/tlv-bei-development-process.

802

Akabane, T., Tabata, K., Kadono, K., Sakuda, S., Terashita, S., Teramura, T., 2010. A Comparison of

803

Pharmacokinetics between Humans and Monkeys. Drug Metab. Dispos. 38, 308–316.

804

doi:10.1124/dmd.109.028829

31

ACCEPTED MANUSCRIPT

805

Batke, M., Escher, S., Hoffmann-Doerr, S., Melber, C., Messinger, H., Mangelsdorf, I., 2011. Evaluation of

806

time extrapolation factors based on the database RepDose. Toxicol. Lett. 205, 122–129.

807

doi:10.1016/j.toxlet.2011.05.1030 Bercu, J.P., Morinello, E., Sehner, C., Shipp, B., Weideman, P., 2016. Point of departure (PoD) selection

RI PT

808 809

for the derivation of acceptable daily exposure (ADE) values for active pharmaceutical ingredients

810

(APIs). Manuscr. Accept. Regul. Toxicol. Pharmacol.

811

Borlak, J., Blickwede, M., Hansen, T., Koch, W., Walles, M., Levsen, K., 2005. Metabolism of verapamil in cultures of rat alveolar epithelial cells and pharmacokinetics after administration by intravenous

813

and inhalation routes. Drug Metab. Dispos. Biol. Fate Chem. 33, 1108–1114.

814

doi:10.1124/dmd.105.003723

M AN U

815

SC

812

Cao, X., Gibbs, S.T., Fang, L., Miller, H.A., Landowski, C.P., Shin, H.-C., Lennernas, H., Zhong, Y.,

816

Amidon, G.L., Yu, L.X., Sun, D., 2006. Why is it challenging to predict intestinal drug absorption

817

and oral bioavailability in human using rat model. Pharm. Res. 23, 1675–1686.

818

doi:10.1007/s11095-006-9041-2

821 822 823

monkey and human. Pharm. Res. 19, 868–874.

TE D

820

Chiou, W.L., Buehler, P.W., 2002. Comparison of oral absorption and bioavailablity of drugs between

Clark, B., Smith, D.A., 1984. Pharmacokinetics and toxicity testing. Crit. Rev. Toxicol. 12, 343–385. doi:10.3109/10408448409044214

Clewell, H.J., Andersen, M.E., Barton, H.A., 2002. A consistent approach for the application of

EP

819

pharmacokinetic modeling in cancer and noncancer risk assessment. Environ. Health Perspect.

825

110, 85–93.

AC C

824

826

Crump, K.S., Chen, C., Chiu, W.A., Louis, T.A., Portier, C.J., Subramaniam, R.P., White, P.D., 2010.

827

What role for biologically based dose-response models in estimating low-dose risk? Environ.

828

Health Perspect. 118, 585–588. doi:10.1289/ehp.0901249

829

Dettli, L., 1982. Pharmacokinetic parameters of interest to the clinician, in: Bolzer, G., van Rossum, J.M.

830

(Eds.), Pharmacokinetics During Drug Development: Data Analysis and Evaluation Techniques.

831

Fischer, Stuttgart, New York, pp. 18– 26.

32

ACCEPTED MANUSCRIPT

832

Dourson, M.L., Felter, S.P., Robinson, D., 1996. Evolution of science-based uncertainty factors in

833

noncancer risk assessment. Regul. Toxicol. Pharmacol. RTP 24, 108–120.

834

doi:10.1006/rtph.1996.0116

836 837

Dourson, M.L., Stara, J.F., 1983. Regulatory history and experimental support of uncertainty (safety) factors. Regul. Toxicol. Pharmacol. RTP 3, 224–238.

RI PT

835

ECHA, 2010. Chapter R.8: Characterisation of dose [concentration]-response for human health., in: Guidance on Information Requirements and Chemical Safety Assessment. Version 2. European

839

Chemicals Agency (ECHA), Helsinki, Finland. Available at: https://echa.europa.eu, Helsinki,

840

Finland.

EFSA, 2012. Guidance on selected default values to be used by the EFSA Scientific Committee,

M AN U

841

SC

838

842

Scientific Panels and Units in the absence of actual measured data. Eur. Food Saf. J. 10, 2579–

843

2611. doi:10.2903/j.efsa.2012.2579

844

EMA, 2014. Guideline on Setting Health Based Exposure Limits for Use in Risk Identification in the Manufacture of Different Medicinal Products in Shared Facilities. EMA/CHMP/ CVMP/

846

SWP/169430/2012. European Medicine Agency (EMA), London. Available at:

847

http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2014/11/WC50017

848

7735.pdf.

849

TE D

845

EMA, 2012. Guideline on the Approach to Establish a Pharmacological ADI. EMA/CVMP/SWP/355689/2006. European Medicine Agency (EMA), London. Available at:

851

http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2012/01/WC50012

852

0832.pdf.

854 855 856 857 858 859

AC C

853

EP

850

Fan, J., Teng, X., Liu, L., Mattaini, K.R., Looper, R.E., Vander Heiden, M.G., Rabinowitz, J.D., 2015. Human phosphoglycerate dehydrogenase produces the oncometabolite D-2-hydroxyglutarate. ACS Chem. Biol. 10, 510–516. doi:10.1021/cb500683c

Gaylor, D.W., 2000. The use of Haber’s law in standard setting and risk assessment. Toxicology 149, 17– 19. Gibaldi, M., Perrier, D. (Eds.), 1982. Pharmacokinetics, Second Edition. CRC Press, New York, pp. 121– 125.

33

ACCEPTED MANUSCRIPT

860

Gould, J.C., Kasichayanula, S., Shepperly, D.C., Boulton, D.W., 2013. Use of low-dose clinical pharmacodynamic and pharmacokinetic data to establish an occupational exposure limit for

862

dapagliflozin, a potent inhibitor of the renal sodium glucose co-transporter 2. Regul. Toxicol.

863

Pharmacol. RTP 67, 89–97. doi:10.1016/j.yrtph.2013.07.002

864

RI PT

861

ICH, 2013. Draft Consensus Guideline. Guideline for Elemental Impurities Q3D. International Conference on Harmonisation (ICH), Geneva. Available at:

866

http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Quality/Q3D/Q3D_Step2

867

b.pdf.

868

SC

865

ICH, 2011. Impurities: Guideline for Residual Solvents Q3C(R5) Step 4 Version. International Conference on Harmonisation (ICH), Geneva. Available at:

870

http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Quality/Q3C/Step4/Q3C_

871

R5_Step4.pdf.

872

M AN U

869

IPCS, 2005. Chemical-specific adjustment factors for interspecies differences and human variability: Guidance document for use of data in dose/concentration-response assessment. International

874

Programme on Chemical Safety (IPCS). World Health Organization (WHO), Geneva. Available at:

875

http://www.inchem.org/documents/harmproj/harmproj/harmproj2.pdf.

876

TE D

873

ISPE, 2010. Baseline Pharmaceutical Engineering Guide, Volume 7, Risk-Based Manufacture of Pharmaceutical Products: A Guide to Managing Risks Associated with Cross Contamination.

878

International Society for Pharmaceutical Engineering (ISPE), Tampa, FL. Available at:

879

http://www.ispe.org/baseline-guides/risk-mapp.

881 882 883 884 885

Kadry, A.M., Skowronski, G.A., Abdel-Rahman, M.S., 1995. Evaluation of the use of uncertainty factors in

AC C

880

EP

877

deriving RfDs for some chlorinated compounds. J. Toxicol. Environ. Health 45, 83–95. doi:10.1080/15287399509531982

Kirman, C.R., Sweeney, L.M., Meek, M.E., Gargas, M.L., 2003. Assessing the dose-dependency of allometric scaling performance using physiologically based pharmacokinetic modeling. Regul Toxicol. Pharmacol. 38(3), 345-67.

34

ACCEPTED MANUSCRIPT

886

Kuempel, E.D., Sweeney, L.M., Morris, J.B., Jarabek, A.M., 2015. Advances in Inhalation Dosimetry

887

Models and Methods for Occupational Risk Assessment and Exposure Limit Derivation. J. Occup.

888

Environ. Hyg. 12 Suppl 1, S18–40. doi:10.1080/15459624.2015.1060328 Labiris, N.R., Dolovich, M.B., 2003. Pulmonary drug delivery. Part II: the role of inhalant delivery devices

890

and drug formulations in therapeutic effectiveness of aerosolized medications. Br. J. Clin.

891

Pharmacol. 56, 600–612.

RI PT

889

Lehman, A.J., Fitzhugh, 0. G., 1954. 100-Fold margin of safety. Assoc. Food Drug US Q. Bull. 18, 33–35.

893

Lewis, S.C., Nessel, C.S., 1994. Extrapolating subchronic test results to estimate chronic no-observed-

895

adverse-effect levels: Factors of 10 are larger than necessary. Toxicologist 14, 401. Lipinski, C.A., Lombardo, F., Dominy, B.W., Feeney, P.J., 2001. Experimental and computational

M AN U

894

SC

892

896

approaches to estimate solubility and permeability in drug discovery and development settings.

897

Adv. Drug Deliv. Rev. 46, 3–26.

898

McNamara, B.P., 1976. Estimates of the toxicity of hydrocyanic acid vapors in man. Edgewood Arsenal Technical Report, EB-TR-76023. Edgewood Arsenal, U.S. Department of the Army, Aberdeen

900

Proving Ground, MD. Available at: http://www.dtic.mil/cgi-

901

bin/GetTRDoc?Location=U2&doc=GetTRDoc.pdf&AD=ADA028501.

TE D

899

Meek, M.E., Renwick, A., Ohanian, E., Dourson, M., Lake, B., Naumann, B.D., Vu, V., 2002. Guidelines

903

for application of chemical-specific adjustment factors in dose/concentration–response

904

assessment. Toxicology 181, 115–120.

906 907 908

Muller, P.Y., Milton, M., Lloyd, P., Sims, J., Brennan, F.R., 2009. The minimum anticipated biological effect level (MABEL) for selection of first human dose in clinical trials with monoclonal antibodies.

AC C

905

EP

902

Curr. Opin. Biotechnol., Chemical biotechnology ● Pharmaceutical biotechnology 20, 722–729. doi:10.1016/j.copbio.2009.10.013

909

Musther, H., Olivares-Morales, A., Hatley, O.J.D., Liu, B., Rostami Hodjegan, A., 2014. Animal versus

910

human oral drug bioavailability: do they correlate? Eur. J. Pharm. Sci. Off. J. Eur. Fed. Pharm.

911

Sci. 57, 280–291. doi:10.1016/j.ejps.2013.08.018

35

ACCEPTED MANUSCRIPT

912

Naumann, B.D., Dolan, D.G., Sargent, E.V., 2004. Rationale for the Chemical-Specific Adjustment

913

Factors Used to Derive an Occupational Exposure Limit for Timolol Maleate. Hum. Ecol. Risk

914

Assess. Int. J. 10, 99–111. doi:10.1080/10807030490280990 Naumann, B.D., Silverman, K.C., Dixit, R., Faria, E.C., Sargent, E.V., 2001. Case Studies of Categorical

RI PT

915 916

Data-Derived Adjustment Factors. Hum. Ecol. Risk Assess. Int. J. 7, 61–105.

917

doi:10.1080/20018091094213

918

Naumann, B.D., Weideman, P.A., 1995. Scientific basis for uncertainty factors used to establish

occupational exposure limits for pharmaceutical active ingredients. Hum. Ecol. Risk Assess. Int.

920

J. 1, 590–613. doi:10.1080/10807039509380049

Naumann, B.D., Weideman, P.A., Dixit, R., Grossman, S.J., Shen, C.F., Sargent, E.V., 1997. Use of

M AN U

921

SC

919

922

toxicokinetic and toxicodynamic data to reduce uncertainties when setting occupational exposure

923

limits for pharmaceuticals. Hum. Ecol. Risk Assess. Int. J. 3, 555–565.

924

doi:10.1080/10807039709383711

925

Naumann, B.D., Weideman, P.A., Sarangapani, R., Hu, S.-C., Dixit, R., Sargent, E.V., 2009. Investigations of the use of bioavailability data to adjust occupational exposure limits for active

927

pharmaceutical ingredients. Toxicol. Sci. Off. J. Soc. Toxicol. 112, 196–210.

928

doi:10.1093/toxsci/kfp195

929

TE D

926

Pfister, T., Dolan, D., Bercu, J., Gould, J., Wang, B., Bechter, R., Barle, E.L., Pfannkuch, F., Flueckiger, A., 2014. Bioavailability of therapeutic proteins by inhalation--worker safety aspects. Ann. Occup.

931

Hyg. 58, 899–911. doi:10.1093/annhyg/meu038

933 934 935 936 937 938

Rennen, M.A.J., Hakkert, B.C., Stevenson, H., Bos, P.M.J., 2001. Data-base derived values for the

AC C

932

EP

930

interspecies extrapolation : a quantitative analysis of historical toxicity data. Comments Toxicol 7, 423–436.

Renwick, A.G., 1993. Data‐derived safety factors for the evaluation of food additives and environmental contaminants. Food Addit. Contam. 10, 275–305. doi:10.1080/02652039309374152

Renwick, A.G., 1991. Safety factors and establishment of acceptable daily intakes. Food Addit. Contam. 8, 135–149. doi:10.1080/02652039109373964

36

ACCEPTED MANUSCRIPT

939

Rhomberg, R.L., Lewandowski, T.A., 2004. Methods for identifying a default cross-species scaling factor.

940

Risk Assessment Forum, U.S. Environmental Protection Agency (EPA), Washington, D.C.

941

Available at: http://www.epa.gov/raf/publications/pdfs/RHOMBERGSPAPER.PDF. RIVM, 2002. Multiple Path Particle Dosimetry Model (MPPD v 1.0): A Model for Human and Rat Airway

943

Particle Dosimetry. RIVA Report 650010030. National Institute for Public Health and the

944

Environment (RIVM), Bilthoven, The Netherlands.

947 948 949

and Febiger, Philadelphia, PA, p. 100.

SC

946

Rowland, M., Tozer, T.N., 1980a. Clinical Pharmacokinetics: Concepts and Applications. 1st Edition. Lea

Rowland, M., Tozer, T.N., 1980b. Clinical Pharmacokinetics: Concepts and Applications. 1st Edition. Lea and Febiger, Philadelphia, PA, pp. 174–179.

M AN U

945

RI PT

942

Sargent, E.V., Faria, E., Pfister, T., Sussman, R.G., 2013. Guidance on the establishment of acceptable

950

daily exposure limits (ADE) to support risk-based manufacture of pharmaceutical products. Regul.

951

Toxicol. Pharmacol. 65, 242–250.

952

Sargent, E.V., Kirk, G.D., 1988. Establishing airborne exposure control limits in the pharmaceutical

953

industry. Am. Ind. Hyg. Assoc. J. 49, 309–313. doi:10.1080/15298668891379792 Sargent, E.V., Naumann, B.D., Dolan, D.G., Faria, E.C., Schulman, L., 2002. The Importance of Human

TE D

954 955

Data in the Establishment of Occupational Exposure Limits. Hum. Ecol. Risk Assess. Int. J. 8,

956

805–822. doi:10.1080/20028091057213

959 960 961 962 963

EP

958

Sharma, V., McNeill, J.H., 2009. To scale or not to scale: the principles of dose extrapolation. Br. J. Pharmacol. 157, 907–921. doi:10.1111/j.1476-5381.2009.00267.x Sietsema, W.K., 1989. The absolute oral bioavailability of selected drugs. Int. J. Clin. Pharmacol. 27,

AC C

957

179–211.

Silverman, K.C., Naumann, B.D., Holder, D.J., Dixit, R., Faria, E.C., Sargent, E.V., Gallo, M.A., 1999. Establishing Data-Derived Adjustment Factors from Published Pharmaceutical Clinical Trial Data. Hum. Ecol. Risk Assess. Int. J. 5, 1059–1089. doi:10.1080/10807039991289347

964

Snodin, D.J., McCrossen, S.D., 2012. Guidelines and pharmacopoeial standards for pharmaceutical

965

impurities: overview and critical assessment. Regul. Toxicol. Pharmacol. RTP 63, 298–312.

966

doi:10.1016/j.yrtph.2012.03.016

37

ACCEPTED MANUSCRIPT

967

Sussman, R.G., Naumann, B.D., Pfister, T., E.P., Sehner, C., Weideman, P., 2016. A harmonization effort

968

for exposure methodology – considerations for application of adjustment factors. Manuscr.

969

Accept. Regul. Toxicol. Pharmacol. Tian, S., Li, Y., Wang, J., Zhang, J., Hou, T., 2011. ADME evaluation in drug discovery. 9. Prediction of

RI PT

970 971

oral bioavailability in humans based on molecular properties and structural fingerprints. Mol.

972

Pharm. 8, 841–851. doi:10.1021/mp100444g

US EPA, 2014. Guidance for Applying Quantitative Data to Develop Data-Derived Extrapolation Factors

974

for Interspecies and Intraspecies Extrapolation. Office of the Science Advisor, Risk Assessment

975

Forum, U.S. Environmental Protection Agency (EPA), Washington, DC. Available at:

976

http://www2.epa.gov/sites/production/files/2015-01/documents/ddef-final.pdf.

M AN U

SC

973

977

US EPA, 2011. Recommended Use of Body Weight ¾ as the Default Method in Derivation of the Oral

978

Reference Dose, EPA/100/R11/0001. Office of the Science Advisor, Risk Assessment Forum,

979

U.S. Environmental Protection Agency (EPA), Washington, DC. Available at:

980

http://www.epa.gov/raf/publications/pdfs/recommended-use-of-bw34.pdf.

981

US FDA, 2008. Guidance for Industry: Genotoxic and Carcinogenic Impurities in Drug Substances and Products: Recommended Approaches. U.S. Food and Drug Administration (FDA), Silver Spring,

983

MD. Available at:

984

http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UC

985

M347725.pdf.

988 989 990 991 992 993 994

EP

987

US FDA, 2005. Guidance for Industry Estimating the Maximum Safe Starting Dose in Initial Clinical Trials for Therapeutics in Adult Healthy Volunteers. U.S. Food and Drug Administration (FDA), Center

AC C

986

TE D

982

for Drug Evaluation and Research, Silver Spring, MD. Available at: http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm 078932.pdf.

Walsh, A., 2011a. Cleaning validation for the 21st century: Acceptance limits for active pharmaceutical ingredients (APIs): Part I. Pharm. Eng. 31, 74–83. Walsh, A., 2011b. Cleaning validation for the 21st century: Acceptance limits for active pharmaceutical ingredients (APIs): Part II. Pharm. Eng. 31, 44–49.

38

ACCEPTED MANUSCRIPT

997 998 999 1000 1001 1002

Acceptance limits for cleaning agents. Pharm. Eng. 12–24. Weil, C.S., McCollister, D.D., 1963. Relationship between short and long term feeding studies in designing an effective toxicity test. Agr. Food Chem 11, 486–491.

RI PT

996

Walsh, A., Ovais, M., Altmann, T., Sargent, E.V., 2013. Cleaning validation for the 21st century:

Welling, P.G., 1995. Differences between pharmacokinetics and toxicokinetics. Toxicol. Pathol. 23, 143– 147.

Woutersen, R.A., Til, H.P., Feron, V.J., 1984. Sub-acute versus sub-chronic oral toxicity study in rats: comparative study of 82 compounds. J. Appl. Toxicol. JAT 4, 277–280.

SC

995

1003 9. Figure Captions

1005

Figure 1. Illustration of the breakdown of the AFH and AFA 10-fold adjustment factors into their TK and TD

1006

components, as suggested by CSAF (IPCS, 2005) or DDEF guidelines (US EPA, 2014). Figure as

1007

adapted from IPCS (2005).

M AN U

1004

1008

Figure 2. Illustration of the time course of oral exposure as it relates to available dose-metrics for use in

1010

CSAF development. Concentration axis is presented on a logarithmic scale axis. Cmax – peak

1011

concentration; AUC – area under the curve; t½ - half-life; Tmax – time to peak concentration.

AC C

EP

TE D

1009

39

ACCEPTED MANUSCRIPT

1

Highlights

2



A number of TK parameters can be used to replace default adjustment factors.

3



TK data can also be used to adjust the point-of-departure (PoD) for intermittent dosing and to

Selection of the best TK parameter requires knowledge of the predictors of target tissue

TE D

M AN U

SC

concentrations and expert judgement.

EP

6



AC C

5

RI PT

account for steady state.

4