throbber
Guidance for Industry
`and Reviewers
`Estimating the Safe Starting Dose in
`Clinical Trials for Therapeutics in
`Adult Healthy Volunteers
`
`DRAFT GUIDANCE
`
`This guidance document is being distributed for comment purposes only.
`
`Comments and suggestions regarding this draft document should be submitted within 60 days of
`publication in the Federal Register of the notice announci ng the availability of the draft
`bruidance Submit comments to Dockets Management Branch (HFA-305), Food and Drug
`Administration, 5630 Fishers Lane, rm. 1061, Rockville, MD 20852. All comments should be
`identified with the docket number listed in the notice of availability that publishes in the Federal
`Register.
`
`For questions regarding this draft document contact (CDER) Robert Osterberg, 30 l-594-5476 or
`(CBER) Martin Green 301-827-5349.
`
`U.S. Dcparhncnt of Health and Human Services
`Food and Drug Administration
`Center for Drug Evaluation and Research (CDER)
`Center for Biologics Evaluation and Research (CBER)
`December 2002
`Pharmacology and Toxicology
`
`hup:llwww.fda.gov/OHRMSIDOCKETSI98fii02d-0492-gdlOOOJ-voll.doc
`09105113
`
`DEFENDANTS' ~
`EXHIBIT
`;
`1
`DTX-1252 ~
`
`DTX-1252-0001
`Dr. Reddy's Laboratories, Ltd., et al.
`v.
`Helsinn Healthcare S.A., et al.
`U.S. Patent No. 8,729,094
`Reddy Exhibit 1031
`
`Exh. 1031
`
`

`
`Guidance for Industry
`and Reviewers
`Estimating the Safe Starting Dose in
`Clinical Trials for Therapeutics in
`Adult Healthy Volunteers
`
`Additional copies are available from:
`
`Office ofTraining and Communications
`Division ofDrug /J~tormafion, HFD-240
`Center for Drug Evaluation and Research
`Food and Drug Administration
`5600 Fishers Lane
`Rockville, MD 20857
`(Fe/) 301-827-4573
`http://www.ji:Ja.govlcderlguidancelindex.htm
`or
`
`Office of Communication, Training and
`Manufacturers Assistance (IJFM-40)
`Center for Biologics Evaluation and Research
`rood and Drug Administration
`1401 Rockville Pike
`Rockville, MD 20852-1448
`(Internet) htlp:llwwwji:Ja.gov!cberlguidelines.htm
`Mail: The Voice Information System a1 800-835-4709 or 301-827-1800
`
`U.S. Department of Health and Human Ser-vices
`Food and Drug Administration
`Center for· Dntg Evaluation and Research (COER)
`Center for Biologics Evaluation and Research (CBER)
`December 2002
`Pharmacology and Toxicology
`
`hltp:/lwww.fda.gowOIIRMS/DOCKETS/98fi102d-0492-gdlOOOJ -vo/ 1 .doc
`I)Y/().">1/J
`
`DTX-1252-0002
`
`Exh. 1031
`
`

`
`Draft - Not for Implementation
`
`Table of Contents
`
`INTRODUCTION ................................................................................................................. 1
`I.
`TJ. SCOPE ................................................................................................................................... 1
`ID. OVERVIEW OF THE ALGORITHM ............................................................................... 3
`IV. STEP l: NO OBSERVED ADVERSE EFFECT LEVEL (NOAEL)
`DETER.MINATION ............................................................................................................ 4
`V. STEP 2: HUMAN EQUIVALENT DOSE (RED) CALCULATION ............................... 5
`A. Conversion Based on Body SurfaceArea .................................................................................... S
`B. Basis for Using Mg/Kg Conversions ............................................................................................. 7
`C. Other Exceptions to Mg/M2 Scaling Between Species ................................................................ 8
`VI. STEP 3: MOST APPROPRIATE SPECIES SELECTION ............................................ 8
`VU. STEP 4: APPLICATION OF SAFETY FACTOR ....................................................... 9
`A.
`Increasing the Safety Facto•· ....................................................................................................... 10
`B. Decreasing the Safety Factor ...................................................................................................... 11
`Vill. STEP 5: CONSIDERATION OF THE PHARMACOLOGICALLY ACTIVE
`DOSE (PAD) ................................................................................................................................ 11
`IX. SUIVIMARY ......................................................................................................................... 12
`REFERENCES ............................................................................................................................ 13
`APPENDIX A .............................................................................................................................. 15
`APPENDIX 8 .............................................................................................................................. 17
`AI~I~ENOIX C .............................................................................................................................. 22
`APPENDIX D .............................................................................................................................. 23
`APPENDIX E .............................................................................................................................. 25
`GLOSSARY ................................................................................................................................. 26
`
`hltp:/lwww.fda.gowOIIRMS/DOCKETS/98fi102d·0492·gdlOOOJ -vo/ 1 .doc
`I)Y/():>1/J
`
`DTX-1252-0003
`
`Exh. 1031
`
`

`
`2
`3
`4
`5
`6
`7
`8
`9
`10
`11
`12
`13
`14
`15
`16
`17
`18
`19
`20
`21
`22
`23
`24
`25
`26
`27
`28
`29
`30
`31
`32
`33
`34
`35
`36
`37
`38
`39
`
`Draft - Not for Implementation
`
`Guidance for Industry and Reviewers'
`
`Estimating the Safe Starting Dose in Clinical Trials
`for Therapeutics in Adult Healthy Volunteers
`
`This draft guidance, when finalized, will represent the Food and Dmg Administration's (FDA's) current
`thinking on this topic. It does not create or confer any rights for or on any person and does not operate to
`bind FDA or the public. An alternative approach may be used if such approach satisfies the requirements
`of the applicable statutes and regulations.
`
`I.
`
`INTRODUCTION
`
`This guidance outlines a process (algorithm) and vocabulary for deriving the maximum
`recommended starting dose (MRSD) for "first in human" clinical trials of new molecular entities
`in adult healthy volunteers and recommends a standardized process by which the MRSD can be
`selected. The purpose of this process is to ensure the safety of the human volunteers.
`
`The goals of this guidance are to (l) establish a consistent terminology for discussing the starting
`dose, (2) provide common conversion factors for deriving a human equivalent dose, and (3)
`delineate a strategy for selecting the rviRSD for adult healthy volunteers, regardless of the
`projected clinical use. This process is diagrammed with a flow chart that presents the decisions
`and calculations used to generate the TYlRSD from animal data.
`
`IT.
`
`SCOPE
`
`The process identified in this document pertains to determining the MRSD for adult healthy
`subjects when beginning a clinical investigation of any new drug or biological therapeutic that
`has been studied in animals. This document is not pertinent to prophylactic vaccines or
`endogenous proteins (i.e., recombinant clottjng factors) used at physiologic concentrations. The
`process outlined in this document does not address dose escalation or maximum allowable doses
`in clinical trials.
`
`1 Tins guidance has been prepared by the Office of New Drugs in the Center for Drug Evaluation and Research
`(CDER) in cooperation with U1e Center for Biologics Evaluation and Research (CBER) at the Food and Drug
`Administration.
`http:llwwwfda.goviOHRMS.fDOCKETSI98.frl02d-0492-f!.dl0001 _,,of 1 .doc
`l
`09105113
`
`DTX-1252-0004
`
`Exh. 1031
`
`

`
`Draft - Not for Implementation
`
`Toxicity should be avoided at the initial dose. However, doses should be chosen that allow
`reasonably rapid attainment of the phase 1 trial objectives (e.g., assessment of the therapeutic's
`tolerability, pharmacodynamic or pharmacokinetic profile). All of the relevant preclinical data,
`including information on the pham1acologically active dose, the full toxicologic profile of the
`compound, and the pharmacokinetics (absorption, distri bution, metabolism, and excretion) of the
`therapeutic, should be considered when determining the MRSD. Starting with doses lower than
`the MRSD is always a possible option and may be particularly appropriate to meet some clinical
`trial objectives.
`
`40 Although the process outlined in this document uses observed toxicities, administered doses, and
`an algorithmic approach to calculate the MRSD, an alternative approach could be proposed that
`41
`42
`places ptimary emphasis on animal phatmacokinetics and modeling rather than dose. In a
`limited number of cases, animal pharmacokinetic data may be useful in determining initial
`43
`44
`clinical doses.Z However, in the majority of new INDs, animal data are not available in
`45
`sufficient detail to construct a scientifically valid, pharmacokinetic model whose aim is to
`46
`accurately project an MRSD.
`47
`48
`49
`50
`51
`52
`53
`54
`55
`56
`57
`58
`59
`60
`61
`62
`63
`64
`65
`
`The remainder of this document will focus on the recommended algorithmic process for starting
`dose extrapolation from animals to humans based on administered doses, since this method will
`likely be useful for the majority of new INDs seeking to investigate new drugs in healthy
`volunteers. Some classes of drugs (e.g., many cytotoxic or biological agents) are commonly
`introduced into initial clinical trials in patient volunteers rather than healthy volunteers.
`Typically, this occurs when a drug is suspected or known to be unavoidably toxic. Although this
`document does not specifically address starting doses in patients, many principles and some
`approaches recommended here may be applicable to designing such trials.
`
`2 If the parent dmg is measured in the plasma at multiple times and fits the range of toxic dose for two or more
`animnl species, it may be possible to develop n phnnnacokinetic model predicting human doses nnd concentrations
`and draw inferences about human safe plasma levels in the absence of prior human data. While quantitative
`modeling for this purpose may be straightforward, tJ1e following points suggest Utis approach may present a number
`of difficulties when evaluating estimates of a safe starting dose. Generally, at the time ofTND initiation, there are a
`number of unknowns regarding animal toxicity and comparability of human and animal phannacokinctics and
`metabolism: ( 1) human bioavailability and metabolism may diller significantly from that of animals; (2)
`mechanisms of toxicity may not be known (i.e., toxic accumulation in a peripheral compartment.; and/or (3) toxicity
`may be due to an unidentified metabolite, not parent drug. Thus, to rely on phannacokinetic models (based on
`parent drug in plasma) to gauge starting doses would require multiple untested assumptions. Modeling may be used
`wiUl greatest validity to estimate human staning doses in special cases where few underlying assumptions would be
`necessary. Such cases are exemplified by large molecular weight proteins (like humanized monoclonaJ antibodies),
`which are intravenously administered, are removed from circulation by endocytosis rather than metabolizism, have
`iuunediate and detectable effects on blood cells, and have a volume of distribution limited to Ule plasma volume.
`Here, allometric, phannacokinetic, and phannacodynamic models have been useful in identifying the human mglkg
`dose that would be predicted to correlate with safe drug plasma levels in nonhuman primates. Even in these cases,
`uncenainties (such as differences between human and chimpanzee receptor sensitivity or density) have been shown
`to affect human phannacologic or toxicologic outcomes, and U1e use of safety factors as described in Ulis docmnent
`is still warranted.
`http:llwwwfda.govi0HRMS/DOCKETSI98.frl02d-0492-f!.dl0001 _,,of 1 .doc
`2
`09105113
`
`DTX-1252-0005
`
`Exh. 1031
`
`

`
`Draft - Not for Implementation
`
`Ill. OVERVIEW OF THE ALGORITHM
`
`66
`67
`68
`The process for selecting the IvfRSD is presented in Figure 1 and described in this section. The
`69
`70 major elements 0 the determination of the no observed adverse effect levels (NOAELs) in the
`71
`tested species, conversion ofNOAELs to human equivalent dose (HED), selection of the most
`appropriate species, and application of a safety factor 0 are aU discussed in greater detail in
`72
`73
`subsequent sections. Situations are also discussed in which the algorithm should be modified.
`The algorithm is intended to be used for systemically administered therapeutics. Topical,
`74
`75
`intranasal, intra-tissue, and compartmental administration routes and depot formulations may
`76
`have additional considerations, but similar principles should apply.
`77
`The process of calculating the MRSD should begin after the toxicity data have been analyzed.
`78
`79 Although only the NOAEL should be used directly in the algorithm for calculating a MRSD,
`80
`other data (exposure/toxicity relationships, pharmacologic data, or prior clinical experience with
`81
`related drugs) can affect the choice of most appropriate species, scaling, and safety factors.
`82
`83
`84
`85
`86
`87
`88
`89
`90
`91
`92
`93
`94
`95
`96
`97 When information indicates that a particular species is most relevant for assessing human risk
`(and deemed the most appropriate species), the HED for that species should be used in
`98
`subsequent calculations, regardless of whether this species was the most sensitive. This case is
`99
`100
`common for biologic therapies, many of which have high selectivity for binding to human target
`proteins, and limited reactivity in species commonly used for toxicity testing. In such cases, in
`101
`102
`vitro binding and activity studies should be done to select appropriate, relevant species before
`toxicity studies are designed (please refer to the ICH3 guidance for industry S6 Prec:linical Safety
`103
`104 Evaluation of Biotechnology-Derived Pharmaceuticals for more details). Additionally, a species
`I 05 might be considered an inapproptiate toxicity model for a given drug if a dose-limiting toxicity
`I 06
`in that species was concluded to be of limited value for human risk assessment (based on
`107
`historical comparisons of toxicities in species to those in humans across a therapeutic class). In
`
`The NOAEL for each species tested should be identified, then each should be converted to the
`human equivalent dose (HED) using appropriate scaling factors. For most systemically
`administered therapeutics, this conversion should be based on the normalization of doses to body
`surface area. Although body surface area conversion is the usual way to approximate equivalent
`exposure if no further information is available, in some cases, extrapolating doses based on other
`parameters may be more appropriate. This decision should be based on the data available for the
`individual case. The body surface area normalization and the extrapolation of the animal dose to
`human dose should be done in one step by dividing the ~OAEL in each of the animal species
`studied by the appropriate body surface area conversion factor (BSA-CF). This is a unitless
`number that converts mg/kg dose for each animal species to the mg/kg dose in humans, which is
`equivalent to the animal's NOAEL on a mg/m2 basis. The resulting figure is called a human
`equivalent dose (HED). The species that generates the lowest HED is called the most sensitive
`spectes.
`
`3 International Conference on Harmonisation of Teclmical Requirements for Registration of Phannaceuticals for
`Human Use (ICH).
`http:llwwwfda.goviOHRMS.fDOCKETSI98.frl02d-0492-f!.dl0001 _,,of 1 .doc
`3
`09105113
`
`DTX-1252-0006
`
`Exh. 1031
`
`

`
`Draft - Not for Implementation
`
`tllis case, data from that species should not be used to derive the HED. Without any additional
`108
`infonnation to guide the choice of the most appropriate species for assessing human risk, the
`109
`I 10 most sensitive species is designated the most appropriate, because using the lowest HED would
`I 11
`generate the most conservative starting dose.
`112
`113 A safety factor should then be applied to the HED to increase assurance that the first dose in
`114
`humans will not cause adverse effects. The use of the safety factor should be based on the
`115
`possibility that humans may be more sensitive to the toxic effects of a therapeutic agent than
`predicted by the animal models, that bioavailability may vary across species, and that the models
`116
`tested do not evaluate all possible human toxicities. For example, ocular disturbances or pain
`117
`118
`(such as severe headaches) in humans can be significant dose-limiting toxicities that may go
`119
`undetected in animal studies.
`120
`In general, a safety factor of 10 is recommended. The MRSD should be obtained by dividing the
`I21
`I 22 HED by the safety factor. Safety concerns or design shortcomings noted in animal studies may
`increase the safety factor, and thus reduce the MRSD further. Alternatively, information about
`I23
`124
`the phannacologic class (well-characterized classes of therapeutics with extensive human clinical
`and preclinical experience) may allay concerns and form the basis of reducing the magnitude of
`I25
`I 26
`the default safety factor and increasing the MRSD. Although a dose lower than the MRSD can
`127
`be used as the actual starting dose, the process described here will derive the maximum
`recommended starting dose. This algorithm generates a MRSD in units ofmg/kg, a common
`128
`129 method of dosing used in phase l trials, but the equations and conversion factors provided in this
`document (Table one, second column) can be used to generate final dosing units in the mg/m2
`130
`131
`form if desired.
`132
`133 As previously stated, for purposes of initial clinical trials in adult healthy volunteers, the HED
`134
`should ordinarily be calculated from the animal NOAEL. If the BED is based on an alternative
`135
`index of effect, such as the pharmacologically active dose (PAD), this exception should be
`136
`prominently stipulated in descriptions of starting dose calculations.
`137
`138
`139
`140
`141
`I42
`143
`STEP 1: NO OBSERVED ADVERSE EFFECT LEVEL (NOAEL)
`IV.
`I44
`I45 DETERMINATION
`146
`147
`148
`I49
`l50
`151
`I 52
`
`The first step in determining the MRSD is to review and evaluate the available animal data so
`that a NOAEL can be determined for each study. Several differing definitions ofNOAEL exist,
`but for selecting a starting dose, the following is used here: the highest dose level that does not
`produce a significant increase in adverse effects. In this context, adverse effects that are
`statistically significant and adverse effects that may be clinically significant (even if they are not
`statistically significant) should be considered in the determination of the NOAEL. The NOAEL
`
`The remainder of this document provides a description of the indi vidual steps in the
`recommended process and the reasoning behind each step. The method is supported by a general
`review and analysis by CDER and CBER examining the results from a number of therapeutics
`entered into development.
`
`http:llwwwfda.goviOHRMS.fDOCKETSI98.frl02d-0492-f!.dl0001 _,,of 1 .doc
`4
`09105113
`
`DTX-1252-0007
`
`Exh. 1031
`
`

`
`153
`154
`155
`156
`157
`158
`159
`160
`161
`162
`163
`164
`165
`166
`167
`168
`169
`170
`I 71
`172
`173
`174
`175
`176
`177
`178
`179
`180
`181
`182
`183
`184
`185
`186
`187
`188
`189
`190
`191
`192
`193
`194
`195
`196
`197
`
`Draft - Not for Implementation
`
`is a generally accepted benchmark for safety when detived from approptiate animal studies and
`can serve as the starting poi nt for determining a reasonably safe starting dose of a new
`therapeutic in healthy (or asymptomatic) human volunteers.
`
`The N OAEL is not the same as the no observed effect level (NOEL), which refers to any effect,
`not just adverse ones, although in some cases the two might be identical. The definition of the
`N OAEL, in contrast to that of the NOEL, reflects the view that some effects observed in the
`animal may be acceptable pharmacodynamic actions of the therapeutic and may not raise a safety
`concern. The NOAEL should not be confused with lowest observed adverse e.ffectlevel
`(LOAEL) or maximum tolerated dose (MID). Both of the latter concepts are based on findings
`of adverse effects and are not generally used as benchmarks for establishing safe starting doses
`in adult healthy volunteers. The tenn level refers to dose or dosage, generally expressed as
`mgfkg or mgfkgfday.
`
`Initial IND submissions for first in human studies by definition lack human data or formal
`allometric comparison of pharmacokinetics. 1-leasurements of systemic levels or exposure (i.e.,
`AUC or Cmax) cannot be employed for setting a safe starting dose in humans, and it is critical to
`rely on dose and observed toxic response data from adequate and well-conducted toxicology
`studies. However, there are cases where data on bioavailability, metabolite profile, and plasma
`drug levels associated with toxicity may influence the choice of the N OAEL. One such case
`would be when saturation of drug absorption occurs at a dose that produces no toxicity. In this
`case, the lowest saturating dose, not the highest (non-toxic) dose, should be used for calculating
`theHED.
`
`There are essentially three types of findings in nonclinical toxicology studies that can be used to
`determine the NOAEL: (1) overt toxicity (e.g., clinical signs, macro- and microscopic lesions);
`(2) surrogate markers of toxicity (e.g., serum liver enzyme levels); and (3) exaggerated
`pharmacodynamic effects. Although the nature and extent of adverse effects can vary greatly
`with different types of therapeutics and it is anticipated that in many instances experts will
`disagree on the characterization of effects as being adverse or not, the use of N OAEL as a
`benchmark for dose-setting in healthy vol unteers should be acceptable to all responsible
`investigators. As a general rule, an adverse effect observed in nonclinical toxicology studies
`used to define a N OAEL for the purpose of dose-setting should be based on an effect that would
`be unacceptable if produced by the initial dose of a therapeutic in a phase l clinical trial
`conducted in adult healthy volunteers.
`
`V.
`
`STEP 2: HUMAN EQUIVALENT DOSE (BED) CALCULATION
`
`A.
`
`Conversion Based on Body Surface Area
`
`After the NOAELs in the relevant animal studies have been determined, they are converted to
`human equivalent doses (HEDs). A decision should be made regarding the most appropri ate
`method for extrapolating the animal dose to the equivalent human dose. Toxic endpoints for
`therapeutics administered systemically to animals, such as the MTD or NOAEL, are usually
`
`http:llwwwfda.goviOHRMS.fDOCKETSI98.frl02d-0492-f!.dl0001 _,,of 1 .doc
`5
`09105113
`
`DTX-1252-0008
`
`Exh. 1031
`
`

`
`198
`199
`200
`201
`202
`203
`204
`205
`206
`207
`208
`209
`2 10
`2 11
`2 12
`2 13
`2 14
`2 15
`2 16
`2 17
`2 18
`2 19
`220
`221
`222
`223
`224
`225
`226
`227
`228
`229
`230
`23 1
`232
`233
`234
`
`Draft - Not for Implementation
`
`assumed to scale well between species when doses are nonnalized to body surface area (i.e.,
`mg/m2
`). The basis for this assumption lies primarily with the work of Freireich et al. (1996) and
`Schein eta!. (1970). These investigators reported that, for antineoplastic drugs, doses lethal to
`10 percent of rodents (LD10s) and MTDs in non-rodents both correlated with the human MTD
`when the doses were normalized to the same administration schedule and expressed as mg/m2.
`Despite the subsequent analyses showing that the MTDs for this set of drugs scale best between
`species when doses are normalized to W0·75 rather than W0
`67 (inherent in body surface area

`normalization), normalization to body surface area has remained a widespread practice for
`estimating an HED based on an animal dose.
`
`An analysis of the impact of the allometric exponent on the conversion of an animal dose to the
`HED was conducted (see Appendix A). Based on this analysis and on the fact that correcting for
`body surface area increases clinical trial safety by resulti ng in a more conservative starting dose
`estimate, it was concluded that the approach of converting NOAEL doses to an HED based on
`body surface area correction factors (i.e., W0 6;) should be maintained for selecting starting doses
`for initial studies in adult healthy volunteers. Nonetheless, use of a different dose normalization
`approach, such as directly equating the human dose to the NOAEL in mg/kg, may be appropriate
`in some circumstances. Deviations from the surface area approach should be justified. The basis
`for justifying direct mg/kg conversion and examples in which other normalization methods are
`appropriate are described in the following subsection.
`
`Although normalization to body surface area is an appropriate method for extrapolating doses
`between species, consistent factors for converting doses from mg/kg to mg/m2 have not always
`been used. Given that body surface area normalization provides a reasonable approach for
`estimating an HED, the factors used for convertin.p doses from each species should be
`standardized. Since surface area varies with W06
`, the conversion factors are therefore
`dependent on the weight of the animals in the studies. However, analyses conducted to address
`the effect of body weight on the actual BSA-CF (body surface area- conversion factor)
`demonstrated that a standard factor provides a reasonable estimate of the HED over a broad
`range of human and animal weights (see Appendix B). The conversion factors and divisors
`shown in Table 1, below, are therefore recommended as the standard val ues to be used for
`interspecies dose conversions for NOAELs in COER and CBER. These factors may also be
`applied when comparing safety margins for other toxicity endpoints (e.g., reproductive toxicity
`and carcinogenicity) when other data for comparison, (i.e., AUCs) are unavailable or are
`otherwise inappropriate for comparison.
`
`Table l: Conversion of Animal Doses to Human Equivalent Doses
`(HED) Based on Body Surface Area
`To convert animal
`To convert animal dose in mg!kg
`to HEDa in mg/kg, either:
`dose in mg/kg to
`dose in mg/m 2,
`Divide
`Multiply
`multiply by km
`animal dose by: Ani mal dose by:
`below:
`
`Species
`
`http:llwwwfda.govi0HRMS/DOCKETSI98.frl02d-0492-gdl0001 _,,of 1 .doc
`6
`09105113
`
`DTX-1252-0009
`
`Exh. 1031
`
`

`
`Draft - Not for Implementation
`
`37
`25
`3
`5
`6
`7
`g
`12
`20
`
`---
`---
`12.3
`7.4
`6.2
`5.3
`4.6
`3.1
`1.8
`
`---
`---
`0.08
`0.13
`0.16
`0.19
`0.22
`0.32
`0.54
`
`Human
`Child (20 kg)b
`Mouse
`Hamster
`Rat
`Ferret
`Guinea pig
`Rabbit
`Dog
`Primates:
`0.32
`3.1
`12
`Monkeys0
`0. 16
`6.2
`6
`Marmoset
`0. 19
`7
`5.3
`Squirrel monkey
`0.54
`20
`1.8
`Baboon
`0.73
`27
`Micro-pig
`14
`0.95
`35
`Mini-pig
`1.1
`a Assumes 60 kg human. For species not Listed or for weights outside the standard ranges, human
`equivalent dose can be calculated from the fonnula:
`HED =animal dose in mglkg x (animal weight in kg/human weight in kg)033.
`b Tllis km is provided for reference only since healthy children will mrely be volunteers for phase 1 trials.
`c For example, cynomolgus, rhesus, stumptaiL
`
`235
`236
`237
`238
`239
`240
`241
`242
`243
`244
`245
`246
`247
`248
`249
`250
`251
`252
`253
`254
`255
`256
`257
`258
`259
`260
`261
`262
`
`B.
`
`Basis for Using Mg/Kg Conversions
`
`The factors in Table I for scaling animal NOAEL to HEDs are based on the assumption that
`doses scale I : 1 between species when normalized to body surface area. However, there are
`occasions for which scaling based on body weight (i.e., setting the HED (mg/kg) = NOAEL
`(mg/kg)) may be more approp1iate. To consider mglkg scaling for a therapeutic, the available
`data should show that the NOAEL occurs at a similar mglkg dose across species. The factors
`below should be satisfied before extrapolating to the RED on a mg/kg basis rather than using the
`mg/m2 approach. Note that mglkg scaling will give a 12-, 6-, and 2- fold higher HED than the
`default mg/m2 approach for mice, rats, and dogs, respectively. If these factors cannot be met, the
`mg/m2 scaling approach for determining the HED should be followed as it will lead to a safer
`MRSD .
`
`1.
`
`2.
`
`NOAELs occur at a similar mg/kg dose across test species (for the studies with a
`given dosing regimen relevant to the proposed initial clinical trial).
`
`If only two NOAELs from toxicology studies in separate species are avai lable,
`one of the following criteria should also be true:
`
`• The therapeutic is administered orally and the dose is limited by local
`toxicities. Gastrointestinal (GI) compartment weight scales by W094
`. GI
`volume determines the concentration of the therapeutic in the GI tract It is
`http:llwwwfda.govi0HRMS/DOCKETSI98_frl02d-0492-f!.dl0001 _,,of 1 .doc
`7
`09105113
`
`DTX-1252-001 0
`
`Exh. 1031
`
`

`
`263
`264
`265
`266
`267
`268
`269
`270
`271
`272
`273
`274
`275
`276
`277
`278
`279
`280
`281
`282
`283
`284
`285
`286
`287
`288
`289
`290
`291
`292
`293
`294
`295
`296
`297
`298
`299
`300
`301
`302
`303
`304
`305
`306
`
`Draft - Not for Implementation
`
`thus reasonable that the toxicity of the therapeutic would scale by mg/kg
`(WLO).
`
`• The toxicity in humans (for a particular class) is dependent on an exposure
`parameter that is highly correlated across species with dose on a mg/kg basis.
`For example, complement activation by systemically administered antisense
`oligonucleotides in humans is believed to be dependent upon Cmax (Geary et
`al., 1997). For some antisense drugs, the Cmax correlates across nonclinical
`species with mg/kg dose and in such instances mg/kg scaling would be
`justified.
`
`• Other pharmacologic and toxicologic endpoints also scale between species by
`mg/kg for the therapeutic. Examples of such endpoints include the MTD,
`lowest lethal dose, and the pharmacologically active dose.
`
`Other Exceptions to Mg/M2 Scaling Between Species
`
`Therapeutics administered by alternative routes (e.g., topical, intranasal,
`subcutaneous, intramuscular) for which the dose is limited by local toxicities.
`Such therapeutics should be normalized to concentration (mg/area of application,
`for instance) or amount of drug (mg) at the application site.
`Therapeutics administered into anatomical compartments that have little
`subsequent distribution outside of the compmtment Examples are intrathecal,
`intravesical, intraocular, intrapleural, and intraperitoneal administration. Such
`therapeutics should be normalized between species according to the
`compartmental volumes and concentrations of the therapeutic.
`Biological products administered intravascularly with Mr > 100,000 daltons. Such
`therapeutics should be normalized to mg/kg.
`
`c.
`1.
`
`2.
`
`-. .> .
`
`VI.
`
`STEP 3: MOST APPROPRIATE SPECIES SELECTION
`
`After the BEDs have been determined from the NOAELs from all toxicology studies relevant to
`the proposed human trial, the next step is to pick one HED for subsequent derivation of the
`MRSD. This HED should be chosen from the most appropriate species. In the absence of data
`on species relevance, a default position is that the most appropriate species for deriving the
`MRSD for a trial in adult healthy volunteers is the most sensitive species (i.e., the species in
`which the lowest HED can be identified).
`
`Factors that could influence the choice of the most appropriate species rather than the default to
`the most sensitive species include: ( l) differences in the absorption, distribution, metabolism and
`elimination (ADME) of the therapeutic between the species; (2) class experience that may
`indicate a particular model is predictive of human toxicity; or (3) limited biological cross-species
`pharmacologic reactivity of the therapeutic. This latter point is especially important for
`
`http:llwwwfda.govi0HRMS/DOCKETSI98,fr102d-0492-f!.dl0001 _,,of 1 .doc
`8
`09105113
`
`DTX-1252-0011
`
`Exh. 1031
`
`

`
`Draft - Not for Implementation
`
`307
`308
`
`biological therapeutics as many are human proteins that bind to human or non-human p1imate
`targets (see ICH guidance S6).
`
`309 When determi ning the MRSD for the first dose of a new therapeutic in humans, absorption,
`310
`distribution, and elimination parameters will not be known for humans. Comparative
`3 11 metabolism data, however, might be available based on in vitro studies. These data are
`312
`particularly relevant when there are marked diffe

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket