throbber
Guidance for Industry
`Estimating the Maximum Safe
`Starting Dose in Initial Clinical Trials
`for Therapeutics in Adult Healthy
`Volunteers
`
`U.S. Department of Health and Human Services
`Food and Drug Administration
`Center for Drug Evaluation and Research (CDER)
`
`July 2005
`Pharmacology and Toxicology
`
`J:\!GUIDANC\5541fnlcln1.doc
`07/06/05
`
`MYLAN - EXHIBIT 1015
`
`

`

`Guidance for Industry
`Estimating the Maximum Safe
`Starting Dose in Initial Clinical Trials
`for Therapeutics in Adult Healthy
`Volunteers
`
`
`
`
`Additional copies are available from:
`
`Office of Training and Communications
`Division of Drug Information, HFD-240
`Center for Drug Evaluation and Research
`Food and Drug Administration
`5600 Fishers Lane
`Rockville, MD 20857
`(Tel) 301-827-4573
`http://www.fda.gov/cder/guidance/index.htm
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`U.S. Department of Health and Human Services
`Food and Drug Administration
`Center for Drug Evaluation and Research (CDER)
`
`July 2005
`Pharmacology and Toxicology
`
`
`
`
`
`
`
`

`

`TABLE OF CONTENTS
`
`
`INTRODUCTION............................................................................................................. 1
`I.
`BACKGROUND ............................................................................................................... 2
`II.
`III. OVERVIEW OF THE ALGORITHM ........................................................................... 3
`IV.
`STEP 1: NO OBSERVED ADVERSE EFFECT LEVEL DETERMINATION........ 5
`V.
`STEP 2: HUMAN EQUIVALENT DOSE CALCULATION...................................... 6
`A. Conversion Based on Body Surface Area ....................................................................................6
`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......................................... 9
`VII. STEP 4: APPLICATION OF SAFETY FACTOR....................................................... 9
`A.
`Increasing the Safety Factor .......................................................................................................10
`B. Decreasing the Safety Factor ......................................................................................................11
`VIII. STEP 5: CONSIDERATION OF THE PHARMACOLOGICALLY ACTIVE
`DOSE................................................................................................................................ 12
`SUMMARY ..................................................................................................................... 12
`IX.
`REFERENCES............................................................................................................................ 13
`GLOSSARY................................................................................................................................. 15
`APPENDIX A:............................................................................................................................. 16
`Analysis of Allometric Exponent on HED Calculations....................................................................16
`APPENDIX B:............................................................................................................................. 18
`Analysis of Body Weight Effects on HED Calculations ....................................................................18
`APPENDIX C:............................................................................................................................. 24
`Derivation of the Interspecies Scaling Factor (Wa/Wh)(1-b)................................................................24
`APPENDIX D:............................................................................................................................. 25
`Examples of Calculations for Converting Animal Doses to Human Equivalent Doses..................25
`APPENDIX E:............................................................................................................................. 27
`Selection of Maximum Recommended Starting Dose for Drugs Administered Systemically to
`Normal Volunteers ...............................................................................................................................27
`
`
`
`
`
`
`
`

`

`
`
`Contains Nonbinding Recommendations
`Guidance for Industry1
`Estimating the Maximum Safe Starting Dose in Initial Clinical
`Trials for Therapeutics in Adult Healthy Volunteers
`
`INTRODUCTION
`
`
`
`
`
`This guidance represents the Food and Drug 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.
`You can use an alternative approach if the approach satisfies the requirements of the applicable statutes
`and regulations. If you want to discuss an alternative approach, contact the FDA staff responsible for
`implementing this guidance. If you cannot identify the appropriate FDA staff, call the appropriate
`number listed on the title page of this guidance.
`
`
`
`
`I.
`
`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: (1) establish a consistent terminology for discussing the
`starting dose; (2) provide common conversion factors for deriving a human equivalent dose
`(HED); and (3) delineate a strategy for selecting the MRSD for adult healthy volunteers,
`regardless of the projected clinical use. This process is depicted in a flow chart that presents the
`decisions and calculations used to generate the MRSD from animal data (see Appendix E).
`
`FDA’s guidance documents, including this guidance, do not establish legally enforceable
`responsibilities. Instead, guidances describe the Agency’s current thinking on a topic and should
`be viewed only as recommendations, unless specific regulatory or statutory requirements are
`cited. The use of the word should in Agency guidances means that something is suggested or
`recommended, but not required.
`
`
`
`
`1 This guidance has been prepared by the Office of New Drugs in the Center for Drug Evaluation and Research
`(CDER) at the Food and Drug Administration.
`
`
`
`1
`
`

`

`Contains Nonbinding Recommendations
`
`BACKGROUND
`
`
`II.
`
`The process identified in this guidance 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 guidance is not pertinent to endogenous hormones and
`proteins (e.g., recombinant clotting factors) used at physiologic concentrations or prophylactic
`vaccines. The process outlined in this guidance pertains primarily to drug products for which
`systemic exposure is intended; it does not address dose escalation or maximum allowable doses
`in clinical trials.
`
`Although the process outlined in this guidance uses administered doses, observed toxicities, and
`an algorithmic approach to calculate the MRSD, an alternative approach could be proposed that
`places primary emphasis on animal pharmacokinetics and modeling rather than dose (Mahmood
`et al. 2003; Reigner and Blesch 2002). In a limited number of cases, animal pharmacokinetic
`data can be useful in determining initial clinical doses.2 However, in the majority of
`investigational new drug applications (INDs), animal data are not available in sufficient detail to
`construct a scientifically valid, pharmacokinetic model whose aim is to accurately project an
`MRSD.
`
`Toxicity should be avoided at the initial clinical 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 pharmacologically active dose, the full toxicologic
`profile of the compound, and the pharmacokinetics (absorption, distribution, metabolism, and
`excretion) of the therapeutic, should be considered when determining the MRSD. Starting with
`doses lower than the MRSD is always an option and can be particularly appropriate to meet some
`clinical trial objectives.
`
`
`
`2 If the parent drug is measured in the plasma at multiple times and is within the range of toxic exposures for two or
`more animal species, it may be possible to develop a pharmacokinetic model predicting human doses and
`concentrations and to draw inferences about safe human plasma levels in the absence of prior human data. Although
`quantitative modeling for this purpose may be straightforward, the following points suggest this approach can
`present a number of difficulties when estimating a safe starting dose. Generally, at the time of IND initiation, there
`are a number of unknowns regarding animal toxicity and comparability of human and animal pharmacokinetics and
`metabolism: (1) human bioavailability and metabolism may differ significantly from that of animals; (2)
`mechanisms of toxicity may not be known (e.g., toxic accumulation in a peripheral compartment); and/or (3)
`toxicity may be due to an unidentified metabolite, not the parent drug. Therefore, relying on pharmacokinetic
`models (based on the parent drug in plasma) to gauge starting doses would require multiple untested assumptions.
`Modeling can be used with greatest validity to estimate human starting doses in special cases where few underlying
`assumptions would be necessary. Such cases are exemplified by large molecular weight proteins (e.g., humanized
`monoclonal antibodies) that are intravenously administered, are removed from circulation by endocytosis rather than
`metabolism, have immediate and detectable effects on blood cells, and have a volume of distribution limited to the
`plasma volume. In these cases, allometric, pharmacokinetic, and pharmacodynamic models have been useful in
`identifying the human mg/kg dose that would be predicted to correlate with safe drug plasma levels in nonhuman
`primates. Even in these cases, uncertainties (such as differences between human and animal receptor sensitivity or
`density) have been shown to affect human pharmacologic or toxicologic outcomes, and the use of safety factors as
`described in this guidance is still warranted.
`
`
`
`2
`
`

`

`Contains Nonbinding Recommendations
`
`
`
`The remainder of this guidance focuses 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 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, patients are
`used instead of healthy volunteers when a drug is suspected or known to be unavoidably toxic.
`This guidance does not address starting doses in patients. However, many principles and some
`approaches recommended here may be applicable to designing such trials.
`
`
`III. OVERVIEW OF THE ALGORITHM
`
`The recommended process for selecting the MRSD is presented in Appendix E and described in
`this section. The major elements (i.e., the determination of the no observed adverse effect levels
`(NOAELs) in the tested animal species, conversion of NOAELs to HED, selection of the most
`appropriate animal species, and application of a safety factor) are all discussed in greater detail in
`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,
`intranasal, intratissue, and compartmental administration routes and depot formulations can have
`additional considerations, but similar principles should apply.
`
`The process of calculating the MRSD should begin after the toxicity data have been analyzed.
`Although only the NOAEL should be used directly in the algorithm for calculating an MRSD,
`other data (exposure/toxicity relationships, pharmacologic data, or prior clinical experience with
`related drugs) can affect the choice of most appropriate species, scaling, and safety factors.
`
`The NOAEL for each species tested should be identified, and then converted to the 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 standard 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 NOAEL in each of the animal species studied by the appropriate body
`surface area conversion factor (BSA-CF). This conversion factor 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 species.
`
`When information indicates that a particular species is more relevant for assessing human risk
`(and deemed the most appropriate species), the HED for that species may be used in subsequent
`calculations, regardless of whether this species is the most sensitive. This situation is more
`applicable to biologic therapies, many of which have high selectivity for binding to human target
`
`
`
`3
`
`

`

`Contains Nonbinding Recommendations
`
`
`proteins and limited reactivity in species commonly used for toxicity testing. In such cases, in
`vitro binding and functional studies should be conducted to select an appropriate, relevant
`species before toxicity studies are designed (refer to ICH guidance for industry S6 Preclinical
`Safety Evaluation of Biotechnology-Derived Pharmaceuticals for more details3). (However, if
`serious toxicities are observed in an animal species considered less relevant, those toxicities
`should be taken into consideration in determining the species to be used to calculate an HED.
`For example, in one particular case, dog was selected as the animal species used for calculation
`of an HED because of unmonitorable cardiac lesions, even though the rat was considered the
`most relevant species based on pharmacological activity data.) Additionally, a species might be
`considered an inappropriate toxicity model for a given drug if the dose-limiting toxicity in that
`species was concluded to be of limited value for human risk assessment, based on historical
`comparisons of toxicities in the animal species to those in humans across a therapeutic class (i.e.,
`the dose-limiting toxicity is species-specific). In this case, data from that species should not be
`used to derive the HED. Without any additional information to guide the choice of the most
`appropriate species for assessing human risk, the most sensitive species is designated the most
`appropriate, because using the lowest HED would generate the most conservative starting dose.
`
` A
`
` safety factor should then be applied to the HED to increase assurance that the first dose in
`humans will not cause adverse effects. The use of the safety factor should be based on the
`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
`tested do not evaluate all possible human toxicities. For example, ocular disturbances or pain
`(e.g., severe headaches) in humans can be significant dose-limiting toxicities that may go
`undetected in animal studies.
`
`In general, one should consider using a safety factor of at least 10. The MRSD should be
`obtained by dividing the 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 the pharmacologic class (well-characterized classes of
`therapeutics with extensive human clinical and preclinical experience) may allay concerns and
`form the basis for reducing the magnitude of the default safety factor and increasing the MRSD.
`Although a dose lower than the MRSD can be used as the actual starting dose, the process
`described in this guidance will derive the maximum recommended starting dose. This algorithm
`generates an MRSD in units of mg/kg, a common method of dosing used in phase 1 trials, but the
`equations and conversion factors provided in this guidance (Table 1, second column) can be used
`to generate final dosing units in the mg/m2 form if desired.
`
`As previously stated, for purposes of initial clinical trials in adult healthy volunteers, the HED
`should ordinarily be calculated from the animal NOAEL. If the HED is based on an alternative
`index of effect, such as the pharmacologically active dose (PAD), this exception should be
`prominently stipulated in descriptions of starting dose calculations.
`
`
`
`3 We update guidances periodically. To make sure you have the most recent version of a guidance, check the CDER
`guidance Web page at http://www.fda.gov/cder/guidance/index.htm.
`
`
`
`4
`
`

`

`Contains Nonbinding Recommendations
`
`STEP 1: NO OBSERVED ADVERSE EFFECT LEVEL DETERMINATION
`
`
`The remainder of this guidance provides a description of the individual steps in the
`recommended process and the reasoning behind each step.
`
`
`IV.
`
`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 definitions of NOAEL exist, but for
`selecting a starting dose, the following is used: the highest dose level that does not produce a
`significant increase in adverse effects in comparison to the control group. In this context,
`adverse effects that are biologically significant (even if they are not statistically significant)
`should be considered in the determination of the NOAEL. The NOAEL is a generally accepted
`benchmark for safety when derived from appropriate animal studies and can serve as the starting
`point for determining a reasonably safe starting dose of a new therapeutic in healthy (or
`asymptomatic) human volunteers.
`
`The NOAEL is not the same as the no observed effect level (NOEL), which refers to any effect,
`not just an adverse one, although in some cases the two might be identical. The definition of the
`NOAEL, 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 also not be confused with lowest observed adverse effect level
`(LOAEL) or maximum tolerated dose (MTD). 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 term level refers to dose or dosage, generally expressed as
`mg/kg or mg/kg/day.)
`
`Initial IND submissions for first-in-human studies by definition lack in vivo human data or
`formal allometric comparison of pharmacokinetics. Measurements 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 nonclinical data on bioavailability,
`metabolite profile, and plasma drug levels associated with toxicity may influence the choice of
`the NOAEL. One such case is when saturation of drug absorption occurs at a dose that produces
`no toxicity. In this instance, the lowest saturating dose, not the highest (nontoxic) dose, should
`be used for calculating the HED.
`
`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 NOAEL as a
`benchmark for dose-setting in healthy volunteers should be acceptable to all responsible
`investigators. As a general rule, an adverse effect observed in nonclinical toxicology studies
`
`
`
`5
`
`

`

`Contains Nonbinding Recommendations
`
`STEP 2: HUMAN EQUIVALENT DOSE CALCULATION
`
`
`used to define a NOAEL 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 1 clinical trial
`conducted in adult healthy volunteers.
`
`
`V.
`
`
`A.
`
`Conversion Based on Body Surface Area
`
`
`After the NOAELs in the relevant animal studies have been determined, they are converted to
`HEDs. A decision should be made regarding the most appropriate method for extrapolating the
`animal dose to the equivalent human dose. Toxic endpoints for therapeutics administered
`systemically to animals, such as the MTD, are usually assumed to scale well between species
`when doses are normalized to body surface area (i.e., mg/m2) (EPA 1992; Lowe and Davis
`1998). The basis for this assumption lies primarily with the work of Freireich et al. (1966) and
`Schein et al. (1970). These investigators reported that, for antineoplastic drugs, doses lethal to
`10 percent of rodents (LD10s) and MTDs in nonrodents 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) (Travis and White 1988; Watanabe et al. 1992), normalization to body surface
`area has remained a widespread practice for estimating an HED based on an animal dose.
`
`An analysis of the affect 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 resulting 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., W0.67) 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 body surface area approach, when describing the
`conversion of animal dose to HED, 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 converting doses for each species should be
`standardized. Since body surface area varies with W0.67, the conversion factors are 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 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 are therefore recommended as the standard
`
`
`
`6
`
`

`

`Contains Nonbinding Recommendations
`
`
`values to be used for interspecies dose conversions for NOAELs. (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.)
`
`
`
`
`Species
`
`Table 1: Conversion of Animal Doses to Human Equivalent Doses
`Based on Body Surface Area
`To Convert
`To Convert Animal Dose in mg/kg
`to HEDa in mg/kg, Either:
`Animal Dose in
`mg/kg to Dose in
`Divide
`Multiply
`mg/m², Multiply
`Animal Dose By
`Animal Dose By
`by km
`
`
`
`---
`---
`37
`Human
`Child (20 kg)b
`---
`---
`25
`0.08
`12.3
`3
`Mouse
`0.13
`7.4
`5
`Hamster
`0.16
`6.2
`6
`Rat
`0.19
`5.3
`7
`Ferret
`0.22
`4.6
`8
`Guinea pig
`0.32
`3.1
`12
`Rabbit
`0.54
`1.8
`20
`Dog
`
`
`
`Primates:
`Monkeysc
`0.32
`3.1
`12
`0.16
`6.2
`6
`Marmoset
`0.19
`5.3
`7
`Squirrel monkey
`0.54
`1.8
`20
`Baboon
`0.73
`1.4
`27
`Micro-pig
`0.95
`1.1
`35
`Mini-pig
`a Assumes 60 kg human. For species not listed or for weights outside the standard ranges,
`HED can be calculated from the following formula:
`HED = animal dose in mg/kg x (animal weight in kg/human weight in kg)0.33.
`b This km value is provided for reference only since healthy children will rarely be volunteers
`for phase 1 trials.
`c For example, cynomolgus, rhesus, and stumptail.
`
`
`B.
`
`Basis for Using mg/kg Conversions
`
`
`The factors in Table 1 for scaling animal NOAEL to HEDs are based on the assumption that
`doses scale 1:1 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 appropriate. To consider mg/kg scaling for a therapeutic, the available
`data should show that the NOAEL occurs at a similar mg/kg dose across species. The following
`circumstances should exist before extrapolating to the HED on a mg/kg basis rather than using
`
`
`
`7
`
`

`

`Contains Nonbinding Recommendations
`
`
`the mg/m2 approach. Note that mg/kg scaling will give a twelve-, six-, and twofold higher HED
`than the default mg/m2 approach for mice, rats, and dogs, respectively. If these circumstances do
`not exist, the mg/m2 scaling approach for determining the HED should be followed as it will lead
`to a safer MRSD.
`
`1. 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). (However, it should be
`noted that similar NOAELs on a mg/kg basis can be obtained across species because of
`differences in bioavailability alone.)
`
`2. If only two NOAELs from toxicology studies in separate species are available, one of the
`following should also be true:
`
` •
`
` The therapeutic is administered orally and the dose is limited by local toxicities.
`Gastrointestinal (GI) compartment weight scales by W0.94 (Mordenti 1986). GI
`volume determines the concentration of the therapeutic in the GI tract. It is then
`reasonable that the toxicity of the therapeutic would scale by mg/kg (W1.0).
`
`• 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.
`
`• There is a robust correlation between plasma drug levels (Cmax and AUC) and dose in
`mg/kg.
`
`
`C.
`
`Other Exceptions to mg/m2 Scaling Between Species
`
`
`Scaling between species based on mg/m2 is not recommended for the following categories of
`therapeutics:
`
`1. 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 (e.g., mg/area of application) or amount of drug (mg) at
`the application site.
`
`2. Therapeutics administered into anatomical compartments that have little subsequent
`distribution outside of the compartment. Examples are intrathecal, intravesical,
`
`
`
`8
`
`

`

`
`
`Contains Nonbinding Recommendations
`
`intraocular, or intrapleural administration. Such therapeutics should be normalized
`between species according to the compartmental volumes and concentrations of the
`therapeutic.
`
`3. Proteins administered intravascularly with Mr > 100,000 daltons. Such therapeutics
`should be normalized to mg/kg.
`
`STEP 3: MOST APPROPRIATE SPECIES SELECTION
`
`
`
`VI.
`
`After the HEDs 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: (1) differences in the absorption, distribution, metabolism,
`and excretion (ADME) of the therapeutic between the species, and (2) class experience that may
`indicate a particular animal model is more predictive of human toxicity. Selection of the most
`appropriate species for certain biological products (e.g., human proteins) involves consideration
`of various factors unique to these products. Factors such as whether an animal species expresses
`relevant receptors or epitopes may affect species selection (refer to ICH guidance for industry S6
`Preclinical Safety Evaluation of Biotechnology-Derived Pharmaceuticals for more details).
`
`When determining the MRSD for the first dose of a new therapeutic in humans, absorption,
`distribution, and elimination parameters will not be known for humans. Comparative
`metabolism data, however, might be available based on in vitro studies. These data are
`particularly relevant when there are marked differences in both the in vivo metabolite profiles
`and HEDs in animals. Class experience implies that previous studies have demonstrated that a
`particular animal model is more appropriate for the assessment of safety for a particular class of
`therapeutics. For example, in the nonclinical safety assessment of the phosphorothioate
`antisense drugs, the monkey is considered the most appropriate species because monkeys
`experience the same dose limiting toxicity as humans (e.g., complement activation) whereas
`rodents do not. For this class of therapeutics, the MRSD would usually be based on the HED for
`the NOAEL in monkeys regardless of whether it was lower than that in rodents, unless unique
`dose limiting toxicities were observed with the new antisense compound in the rodent species.
`
`
`VII. STEP 4: APPLICATION OF SAFETY FACTOR
`
`Once the HED of the NOAEL in the most appropriate species has been determined, a safety
`factor should then be applied to provide a margin of safety for protection of human subjects
`receiving the initial clinical dose. This safety factor allows for variabil

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