throbber
Eur J Clin Pharmacol (2002) 57: 835–845
`DOI 10.1007/s00228-001-0405-6
`
`R EV IE W A RT I C L E
`
`Bruno G. Reigner Æ Karen Smith Blesch
`Estimating the starting dose for entry into humans:
`principles and practice
`
`Received: 27 August 2001 / Accepted in revised form: 5 November 2001 / Published online: 29 January 2002
`Ó Springer-Verlag 2002
`
`Abstract Background: Selection of the starting dose for
`the entry into humans (EIH) study is an essential first
`step in clinical drug development.
`Objectives: This paper is a review of different approaches
`that may be used to calculate the starting dose, presents
`the results of a current practice survey that reflect
`practice patterns at a large pharmaceutical company,
`and discusses selected topics related to the calculation of
`the starting dose.
`Results: The methods used in the field of oncology for
`cytotoxic compounds are usually derived from a dose
`associated with some toxicity in animals multiplied by a
`safety factor. In therapeutic areas other than oncology,
`the methods may be
`classified as
`four different
`approaches: (1) dose by factor methods that utilize the
`no observable adverse effect level (NOAEL) from pre-
`clinical toxicology studies multiplied by a safety factor;
`(2) the similar drug approach that may be used when
`clinical data are available for another compound of the
`same chemical class as the investigational drug; (3) the
`pharmacokinetically guided approach that uses systemic
`exposure rather than dose for the extrapolation from
`animal to man; and (4) the comparative approach that
`consists of utilizing two or more methods to estimate a
`starting dose and then critically comparing the results to
`arrive at the optimal starting dose. A ‘‘real-life’’ example
`illustrates the use of each method. Advantages, limita-
`tions, and underlying assumptions of each of
`the
`
`B.G. Reigner (&)
`Pharma Development, Clinical Science,
`Clinical Pharmacology Science (52/1010),
`F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124,
`4070 Basel, Switzerland
`E-mail: bruno.reigner@roche.com
`Tel.: +41-61-6884507
`Fax: +41-61-6881434
`
`K.S. Blesch
`Pharma Development,
`F. Hoffmann-La Roche Inc.,
`Nutley, USA
`
`methods are discussed. The results of the survey showed
`that the pharmacokinetically guided approach is the
`most commonly used method, followed by dose by
`factor methods.
`Conclusion: The task of estimating the starting dose is
`moving beyond empirical methods to those that are in-
`creasingly more systematic and theory based.
`
`Keywords Starting dose Æ Entry into human study Æ
`Pharmacokinetics
`
`Introduction
`
`The entry into humans (EIH) study is the first step in the
`clinical development of any molecule that has shown
`therapeutic promise in preclinical evaluations. An
`essential element of the EIH (also known as ‘‘entry into
`man, EIM’’ or ‘‘first time in man, FTIM’’) study is the
`calculation of the starting dose. Estimating the starting
`dose is a very common and important task, yet there is
`little uniformity or
`standardization of approaches.
`Starting-dose calculations are performed in many
`different ways, very often using empirical methods.
`The approach used often depends on the training and
`experience of the scientists involved, and/or the indus-
`trial or academic setting. Individual scientists may have
`their own rules and methods. Occasionally there is
`some consistency within a pharmaceutical company or
`academic setting, but the methods used vary consider-
`ably across these institutions.
`Estimating the optimal starting dose is complicated
`and presents new challenges each time it
`is done.
`Extrapolation of doses from animals to humans is
`based on multiple assumptions about the compound’s
`behavior across species. Different methods may yield
`widely varying results, and an approach that has
`worked well for one compound may not be appropriate
`for another compound. It is important to find a starting
`dose that is low enough to be safe in humans, but not
`so conservative that excessive costly and time-consum-
`
`CFAD Exhibit 1044
`
`

`
`836
`
`ing dose escalations are needed. These challenges make
`it difficult to establish standard rules for this under-
`taking.
`In 1981 it was noted that
`considerable
`uncertainty and controversy surround the choice of the
`initial human dose of a drug [1], a circumstance that
`was echoed in 1990 [2]. More recently, Boxenbaum and
`DiLea [3] argued that, while there were some basic rules
`that could be followed to ensure the safety of patients
`and healthy volunteers, the present state of knowledge
`did not allow development of realistic or reasonable
`standardized procedures for determining optimal start-
`ing doses for entry into humans.
`These sentiments seem to be confirmed by the wide
`variability in approaches used for starting-dose estima-
`tion and the fact that there currently are no guidance
`documents available from regulatory health authorities
`on this topic. A literature search identified a few papers
`that broadly address the issue of starting dose, most
`from the 1970s and 1980s [1, 2, 4, 5, 6] in addition to the
`1995 paper [3] cited above. The number of references is
`small
`considering that a starting-dose
`calculation
`is needed for each molecule early in drug development –
`a very large number. This is possibly a reflection of the
`complexity of the task and the complicated but incom-
`plete knowledge base that underlies it. The objectives of
`this article are to review the different approaches used to
`calculate the starting dose, to illustrate the approaches
`with a ‘‘real-life’’ example, to present the results of a
`current practice survey that reflect actual practice
`patterns at a large pharmaceutical company, and to
`discuss selected topics related to the calculation of the
`starting dose.
`
`General considerations
`
`For non-cytotoxic compounds, the initial EIH study is
`usually a single ascending dose (SAD) study in healthy
`volunteers. The main purpose of the study is to assess
`the tolerability of the new compound after administra-
`tion of a single dose and to gain some information about
`the pharmacokinetics and pharmacodynamics (if possi-
`ble) of the compound in human subjects. Dose estima-
`tion is based on a dose found to be safe in preclinical
`studies and then adjusted for human use, using various
`correction factors to ensure human safety. The optimal
`EIH starting dose is one that is safe, and not pharma-
`codynamically active, but is close to a dose with some
`minimal pharmacodynamic effect in humans. A starting
`dose that is too low results in the expenditure of addi-
`tional time and resources in reaching potential infor-
`mative and therapeutic dose levels, while a starting dose
`that is too high compromises subject safety and may
`overlook important clinical considerations for lower
`doses.
`The approach just described is generally applicable
`to compounds
`in all
`therapeutic areas with the
`exception of cytotoxic compounds intended to treat
`malignant disease. EIH studies for cytotoxic agents are
`
`conducted in patients with treatment-refractory cancer
`instead of healthy volunteers. In EIH studies for
`antineoplastics, there is always hope for a therapeutic
`benefit and a desire to minimize patient exposure to
`sub-therapeutic doses [7, 8], therefore the EIH study
`for these compounds is usually a multiple ascending
`dose (MAD) rather than a SAD study. In general,
`cytotoxic compounds have a very low therapeutic in-
`dex and a steep concentration–response curve for
`safety; however, in oncology there is considerably more
`acceptance of toxicity to achieve therapeutic benefit.
`The starting-dose calculation for antineoplastics
`is
`generally based on a dose and dose schedule that have
`elicited some toxicity in animals rather than on a dose
`that has been identified as safe in animals. Starting
`doses of anti-cancer agents have traditionally been
`established with the goal of escalating quickly to a
`maximum tolerated dose (MTD) on a given dosing
`schedule.
`There are multiple preclinical doses that can serve
`as the basis for estimating the starting dose. They
`have nearly identical meanings and are occasionally
`used interchangeably, although some subtle differences
`exist. For
`example,
`the highest non-toxic dose
`(HNTD) originated in cancer research and is defined
`as
`the highest dose at which no hematological,
`chemical,
`clinical, or morphological drug-induced
`alterations occur, and doubling this dose produces the
`aforementioned alterations
`[9]. The no observable
`adverse effect level (NOAEL) is defined as the highest
`dose at which no statistically significant and/or
`biologically relevant adverse effect is observed [10].
`For the purposes of this paper we will use the term
`NOAEL when discussing doses from preclinical toxi-
`cology studies.
`Figure 1 describes a simple decision tree for esti-
`mating a starting dose and serves as an organizing
`framework for this paper. Methods for estimating the
`starting dose are different depending on whether or not
`the drug is a cytotoxic intended for antineoplastic
`purposes; we have used this basic principle to divide
`the paper into its two main sections. If a drug is not a
`cytotoxic intended for antineoplastic purposes then,
`after estimating a starting dose on the basis of the
`NOAEL, it is reasonable to consider whether or not
`this dose is expected to be pharmacodynamically active
`and needs to be adjusted downward for entry into
`man.
`
`Methods used for cytotoxic compounds
`
`The literature related to estimating a starting dose for
`antineoplastics
`suggests an organized, knowledge-
`building effort by scientists focused on the task, and a
`clear evolution in methods is seen [11, 12, 13, 14, 15, 16],
`although considerable uncertainty continues to exist in
`this therapeutic area. Historically, starting doses of
`antineoplastics for humans were first extrapolated from
`
`

`
`Fig. 1. Decision tree for start-
`ing-dose calculation
`
`837
`
`toxic doses determined in large animal species (e.g., dog
`and monkey). In 1979, based on a retrospective analysis
`of 12 anti-tumor agents, Penta and colleagues [14]
`demonstrated that mouse data could be effectively used
`in determining safe starting doses. In 1981, mouse data
`were further validated against traditional large-animal
`methods with 21 antineoplastic agents and were also
`found to produce safe starting doses [15]. Methods
`based on large animal species and on mice are both in
`use today.
`
`One-third of the toxic dose low in a large animal species
`
`Using this approach, the starting dose is calculated
`as one-third of the toxic dose low (TDL; expressed as
`mg/m2) in a large animal species (either dog or monkey).
`This method was initially introduced by Freireich and
`colleagues [11] and remains widely in use. TDL is
`defined as the lowest dose that produces drug-induced
`pathological alterations
`in hematological, chemical,
`clinical, or morphological parameters and which, when
`doubled, produces no lethality [9]. The TDL is deter-
`mined on two basic schedules, single dose and daily for
`5 days [14].
`
`One-tenth of the lethal dose in mice
`
`Here the starting dose for EIH is calculated as one-
`tenth of a dose (expressed in mg/m2) that is lethal to
`10% of non-tumor bearing mice (LD10) during a
`specified period of observation [14]. The LD10 is de-
`termined on two basic schedules (single dose and daily
`for 5 days) with groups of ten mice at each dose level
`[14]. Collins et al.
`[17]
`investigated the use of 1/10
`LD10 and noted that, for eight drugs, the area under
`the plasma concentration–time curve (AUC) of the
`compound observed in mice after administration of
`the LD10 was similar to that of the compound pro-
`duced by the MTD in humans. The use of a new
`approach to dose escalation called pharmacologically
`guided dose escalation (PGDE) was advocated [18].
`PGDE is now a well-accepted element of oncology
`phase-1 study design. Most papers that address issues
`related to PGDE utilize 1/10 LD10 in mice as the
`method of reference for estimating the starting dose
`[18, 19, 20].
`If
`there is a significant discrepancy
`between 1/10 LD10 in mice and 1/3 TDL in large
`species, other authors have suggested that the lower
`of
`the two doses be used as the starting dose in
`conjunction with PGDE.
`
`

`
`838
`
`Example
`
`For each method described in this paper, a ‘‘real-life’’
`example of a starting-dose calculation is given. For the
`examples, we use the compound mofarotene. Mofaro-
`tene (Ro 40-8757) is a retinoid with cytostatic properties
`that was used in phase-I clinical trials for a potential
`antineoplastic indication. Mofarotene was chosen for
`the example because several methods for estimating the
`starting dose were used and compared before EIH,
`including methods that are usually restricted to cyto-
`toxic drugs. Preclinical studies had indicated a NOAEL
`of 2 mg/kg/day in dogs and 50 mg/kg/day in rats. The
`(95 mg/m2/day).
`TDL in dogs was
`5 mg/kg/day
`The LD10 in mice was not available. Using the 1/3 TDL
`in large animal species, the estimated starting dose for
`mofarotene was:
`1=3  95 mg=m2  1:8 m2 ¼ 57 mg
`ð1Þ
`where 1.8 is the average body surface area of a human in
`m2. The use of a fixed single dose in this example is an
`exception for an oncology phase-I trial. Generally, for
`cytotoxics, the starting dose for EIH studies in cancer
`patients is individualized based on patient body surface
`area rather than using a fixed dose for all patients [21].
`Because mofarotene is cytostatic, with considerably less
`toxicity than a cytotoxic drug, it was initially adminis-
`tered as a single dose to healthy volunteers.
`
`Critical assessment of the method
`
`Multiple variations of the two basic approaches have
`been used by individual investigators and groups for
`various drugs (e.g., 1/50 safe dose in mouse for faz-
`arabine, 1/3 dog TDL for elsamitrucin and docetaxel,
`1/20 lethal dose in rat for gemcitabine, and less than
`1/10 mouse lethal dose for trimetrexate, [22]). Despite
`the substantial work that has been done with starting
`doses for cytotoxics, there is still considerable vari-
`ability, the basic methods haven’t changed, and there
`is no ‘‘gold standard’’ for estimating the starting dose
`for oncology phase-I clinical trials. There has been
`wide concern that the basic methods provide doses
`that are too conservative and miss opportunities for
`therapeutic benefit in oncology phase-I and -II clinical
`trials [8, 16]. PGDE is considered to be an essential
`element of effective phase-I study design in this thera-
`peutic area.
`
`Methods used for non-cytotoxic compounds
`
`There are four basic approaches to estimating the
`starting dose for EIH studies for non-cytotoxic com-
`pounds. For the purposes of this paper they are called:
`dose by factor, similar drug, pharmacokinetically guid-
`ed, and comparative. The comparative approach uses
`data obtained from the other three approaches to criti-
`
`cally evaluate and determine which starting dose is
`optimal. We describe each approach,
`including its
`strengths and weaknesses, and variations that have
`evolved from the original.
`
`The dose by factor approach
`
`This method consists of identifying a dose (usually
`expressed in mg/kg/day) associated with a specific effect
`in preclinical toxicology studies and then multiplying it
`by one or more factors to estimate a safe human starting
`dose. A commonly used approach is based on the
`highest dose of the compound found to have no toxic
`effect in the most sensitive species tested in 4-week to 13-
`week preclinical toxicology studies. This mg/kg/day dose
`is then reduced by a ‘‘sensitivity’’ factor that adjusts for
`anticipated differences in sensitivity to the drug between
`each animal species tested and man. The sensitivity
`factor is derived from estimated interspecies differences
`in sensitivity to drug toxicity published by the Associa-
`tion of Food and Drug Officials (AFDO) of the United
`States in 1959 [23]. According to this ‘‘modified AFDO’’
`scheme, the maximum starting dose for the EIH study is
`the smallest of the following three doses: 1/10 of the
`highest no-effect dose in rodents, 1/6 of the highest
`no-effect dose in dogs, or 1/3 of the highest no-effect
`dose in monkeys [3, 24]. The smallest of the three doses
`is utilized because it reflects which animal species is most
`sensitive to the drug from the toxicology studies. The
`sensitivity factor (i.e., 1/10, 1/6, 1/3) reflects anticipated
`differences in drug sensitivity in the various animal
`species relative to humans [23]. If, for some reason, there
`is concern about the safety of the starting dose derived
`this way, the dose can be further reduced using an
`arbitrary safety factor.
`
`Example
`
`For mofarotene, the dog was the most sensitive species,
`with a NOAEL of 2 mg/kg/day. The starting dose was
`estimated using the ‘‘modified AFDO’’ approach as:
`ð2Þ
`1=6  2mg=kg  70kg ¼ 23mg
`where the NOAEL in the dog was multiplied by 1/6 and
`then 70 kg (average body weight of a human) to estimate
`a starting dose of 23 mg. Because the toxicologist was
`concerned about skin toxicity appearing several weeks
`after the start of administration, the 23-mg dose estimate
`was multiplied by a safety factor of 1/10 to give a final
`starting dose of 2.3 mg.
`
`Critical assessment of the method
`
`There are multiple variations of this approach that
`allow for more or less conservative results. Different
`sensitivity or
`safety factors may be applied. For
`
`

`
`example, a sensitivity factor of 1/2 rather than 1/3 has
`been suggested when extrapolating from primates [25].
`Kuhlman [26] noted that generally the starting dose is
`about 1/50 to 1/100 or lower of the no-effect dose from
`toxicology studies.
`This is a classic approach that has been widely used
`and generally produces safe starting doses for EIH
`studies. Dose by factor methods have been criticized
`because they ignore preclinical pharmacokinetic data [3,
`24]. The approach is somewhat simplistic and empirical,
`and easily lends itself to variations that may be consid-
`ered to be rather arbitrary in nature. It has been noted
`that this type of extrapolation from animals to humans
`is truly appropriate only if both show similar absorp-
`tion, bioavailability, biotransformation, and sensitivity
`to toxic effects by the drug or its biotransformation
`products [25]. While there is considerable flexibility and
`good safety with results, dose by factor methods may be
`criticized for estimating starting doses that are too
`conservative, requiring excessive dose escalations to
`reach a pharmacodynamically active or maximum tol-
`erated dose.
`
`Similar drug approach
`
`The similar drug approach is used when human safety
`data are available for a drug similar to the one under
`investigation and can serve as a reference point for
`estimating the starting dose [1, 5]. The ‘‘similar drug’’
`is usually of the same chemical class, with similar or
`related chemical
`structure. This
`situation is not
`uncommon in industry where one or more predecessor
`compounds in the same chemical class and with sim-
`ilar toxicological profiles may have been clinically
`investigated prior to the current drug under investi-
`gation. The ‘‘similar drug’’
`is one that
`is already
`marketed or that has clinical safety data available
`when the compound under investigation is a follow-up
`compound.
`This approach is based on the ratio of an optimal
`starting dose of the similar drug to its NOAEL. This
`optimal starting dose is one that has been identified as
`producing no drug-related adverse events or laboratory
`abnormalities after a single dose in humans and with no
`pharmacodynamic activity. The method assumes that
`this ratio is equal to the ratio of the starting dose for the
`compound under investigation to its NOAEL. The
`assumption can be expressed as:
`SDs=NOAELs ¼ SDi=NOAELi
`where:
`• SDs is the optimal starting dose of the similar drug.
`• NOAEL is the no-observable adverse effect level for
`drugs ‘‘s’’ and ‘‘i’’, where ‘‘s’’ is the similar drug and
`‘‘i’’ is the investigational drug.
`• SDi is the estimated starting dose for the investiga-
`tional drug.
`
`ð3Þ
`
`839
`
`The ratio SDs/NOAELs can then be applied to the
`NOAELi to estimate a starting dose that is expected to
`be safe, but not too conservative as:
`ð4Þ
`SDi ¼ ðSDs=NOAELsÞ  NOAELi
`The dose estimate obtained this way is usually multiplied
`by an arbitrary safety factor to accommodate uncer-
`tainty about safety in the estimate of the starting dose.
`
`Example
`
`For mofarotene, the similar drug was etretinate, a
`closely related retinoid with a similar toxicity profile in
`animals; the dose that would be an optimal starting dose
`of etretinate in humans was 10 mg. The NOAEL of
`etretinate in rats was 2 mg/kg/day and that of mofaro-
`tene in rats was 50 mg/kg/day. The NOAEL of etreti-
`nate in dogs was unknown. Applying Eq. 4:
`SDi¼ð10mg=2mg=kg70kgÞ50mg=kg70kg¼250mg:
`ð5Þ
`This dose was then multiplied by a safety factor of 1/4 to
`give a final human starting dose of 63 mg. In this case, as
`with the dose by factor method, an arbitrary safety
`factor was applied to ensure the safety of the healthy
`volunteers.
`
`Critical assessment of the method
`
`This ‘‘similar drug’’ method is not new, makes intuitive
`sense, and is known to provide safe starting doses. The
`main limitation is that applying a cross-species dosing
`ratio for one drug to another drug assumes that phar-
`macokinetic and pharmacodynamic differences between
`animal and man are the same for both compounds. The
`validity of this assumption should always be considered
`and may be tested by calculating the ratio SDs/NOAELs
`for another similar drug of the same chemical class to
`verify that the ratio remains reasonably constant. If this
`second ratio is not similar to the first, this approach
`should not be used.
`
`Pharmacokinetically guided approach
`
`The pharmacokinetically guided approach is increas-
`ingly being used in many pharmaceutical companies and
`institutions [24]. It uses systemic exposure instead of
`dose for the extrapolation from animal to man, a con-
`cept that was originally proposed more than three
`decades ago [26]. A desired systemic exposure (e.g.,
`AUC) for humans is defined as the systemic exposure
`corresponding to the NOAEL. If a NOAEL and its
`corresponding AUC are available from more than one
`animal species, the animal species with the lowest AUC
`is used. The clearance of the drug in humans (CLh) is
`
`

`
`840
`
`predicted using allometric scaling [27, 28, 29]. The
`starting dose, with AUC as the measure of systemic
`exposure, is then calculated as:
`SD ¼ AUCp  CLh
`ð6Þ
`where SD is the starting dose, AUCp is the AUC
`obtained at the NOAEL in preclinical toxicology stud-
`ies, and CLh is the clearance in humans predicted by
`allometric scaling.
`The correlation between pharmacokinetics and tox-
`icity in preclinical toxicology studies may be better with
`maximum concentration (Cmax) than AUC. In this case,
`Cmax rather than AUC should be used as the measure of
`systemic exposure. A detailed description of methods for
`and results of interspecies scaling as it applies to dose
`selection for humans is available for a new glycine
`antagonist [30].
`
`Example
`
`For mofarotene, the AUC in dogs was 17.3 lg h/ml at
`the NOAEL. Using allometric scaling and in vivo
`pharmacokinetic data from three animal species (mouse,
`rat, and dog), as well as intrinsic clearance data from
`mouse, rat, dog, and human microsomes, the predicted
`CLh was 16.0 l/h [31]. Applying Eq. 4, the calculated
`starting dose was:
`ð7Þ
`17:3lg  h=mL  16:0L=h ¼ 277mg
`This dose was then multiplied by a safety factor of 1/3,
`which gave a final starting-dose estimate of 92 mg.
`
`Critical assessment of the method
`
`This method also provides safe starting doses [24]. The
`main limitations concern assumptions about interspecies
`differences in concentration–effect relationships and
`metabolism. Defining a systemic exposure for humans
`on the basis of preclinical data assumes that the con-
`centration–effect relationship for the drug is the same in
`animal and man. This assumption should always
`be kept in mind because interspecies differences in
`concentration–effect relationships do exist
`for some
`pharmacological classes such as cardiac glycosides [32].
`If there are concerns about differences in concentra-
`tion–effect relationships between animal and man, it is
`important to quantify them whenever possible. With
`certain compounds, this may be accomplished with in
`vitro testing. For example, for a platelet aggregation
`inhibitor, human and animal platelets can be tested with
`the drug to determine the concentration–response curve
`and evaluate any pharmacodynamic differences between
`animal and man.
`If differences in the concentration–effect relationships
`can be quantified, then this information can be incor-
`porated into the calculations. As an example, assume
`
`that in-vitro testing determines that the maximum effect
`of a compound is the same in animals and humans and
`that the ratio EC50 human/EC50 animal =0.1, where
`EC50 is the drug concentration that elicits 50% of the
`maximum effect. In this case, the drug is ten times more
`potent in man than in animal, and the starting dose for
`man should then be reduced to 1/10 of that originally
`estimated. The method then becomes a pharmacoki-
`netically and pharmacodynamically guided calculation
`of the starting dose.
`this pharmacokinetically
`Another shortcoming of
`guided approach is uncertainty related to the prediction
`of CLh using allometric scaling. Allometric scaling is a
`challenging scientific area and there is always uncer-
`tainty surrounding the prediction of clearance in
`humans [24, 29]. Standard methods are known to work
`well for drugs that are eliminated primarily by renal
`excretion [33, 34]. Allometric scaling methods are less
`reliable for drugs that are primarily eliminated via
`hepatic metabolism. However, recent advances in this
`field suggest that clearance in animals can be corrected
`using intrinsic clearances obtained in vitro with human
`and animal hepatocytes [27]. Using allometric scaling
`combining in vitro and in vivo data from three animal
`species and in vitro data from humans, a retrospective
`analysis of a large number of drugs showed that the
`prediction of CLh was successful in 82% of the cases;
`that is, predictions were within a twofold factor of actual
`human clearance values [27]. For mofarotene, the mea-
`sured CLh was 31.5 l/h [31], slightly less than a twofold
`difference between the predicted and measured CLh. The
`uncertainty associated with allometric scaling should be
`kept in mind when using the pharmacokinetically guided
`approach. As with the other methods described, an
`arbitrary safety factor may be applied to adjust the
`starting dose on the basis of the technique used for
`allometric scaling and its degree of reliability.
`The pharmacokinetically guided approach carries
`additional complexities that must be considered. The
`first concerns which plasma drug concentration is
`appropriate to use in the calculations. When deriving
`measures of systemic exposure from plasma concentra-
`tion data for the purposes described here, it is important
`to consider plasma concentrations of unbound drug
`(Cu). Cu is responsible for its pharmacodynamic effects,
`and Cu in plasma is expected to be more closely related
`to Cu at the site of action than the total plasma
`concentration. It is well established that the unbound
`fraction (fu where fu=Cu/total plasma concentration)
`can vary between species [32, 35]. Therefore, using Cu in
`the calculations of systemic exposure (AUC or Cmax) is
`highly recommended, especially for drugs that are highly
`protein bound in plasma. A striking example of inter-
`species differences in plasma protein binding is etopo-
`side, with unbound fractions of 0.63, 0.52, and 0.048 in
`dog, rat, and man, respectively [32]. In this case, relying
`on total plasma concentration rather than Cu for
`calculating the desired systemic exposure could result in
`an unnecessarily conservative starting dose.
`
`

`
`Other complexities of the pharmacokinetically guided
`approach concern the dose linearity of the drug’s phar-
`macokinetics and its metabolism. We describe this
`approach in the context of linear pharmacokinetics and
`with the parent drug being the sole active moiety.
`Reality, of course can be more complex, for example,
`with non-linear pharmacokinetics and/or the presence of
`active metabolites that must be considered in the start-
`ing-dose calculation. Another issue may involve the use
`of different formulations in animal and human studies
`(e.g., drinking solution in animals and tablet in man).
`When experimental data are available for systemic
`exposure to active metabolites or the relative bioavail-
`ability of the formulations, this information should also
`be included in the starting-dose calculation. It is beyond
`the scope of this paper to describe the integration of this
`information into the starting-dose estimate because it
`will differ for each compound; however, the general
`principles
`regarding pharmacokinetics,
`interspecies
`scaling, and pharmacodynamics will apply.
`
`The comparative approach
`
`This approach consists of estimating the starting dose
`using at least two but preferably three or four methods
`and then comparing the results and interpreting the
`differences to arrive at an optimal starting dose. To
`illustrate this approach, we utilize the starting doses
`estimated for mofarotene in the examples (Table 1).
`For mofarotene,
`three different approaches (1/3
`TDL, similar drug, pharmacokinetically guided) gave
`approximately similar results (57–92 mg), while the dose
`by factor approach with a safety factor of 1/10 gave a
`considerably lower estimate (2.3 mg). Such results are
`common. Methods based on a dose by factor approach
`usually provide a much lower starting-dose estimate
`than other methods. With mofarotene, a starting dose of
`50 mg was selected for the EIH study in healthy vol-
`unteers. This dose was selected because it was felt that
`the 2.3-mg starting dose from the ‘‘dose by factor’’
`approach was too conservative, and 50 mg was at
`the conservative end of the dosing range indicated by the
`other three methods. Results of the EIH study showed
`that the starting dose and a dose of 150 mg were safe (no
`drug-related adverse event or laboratory abnormalities)
`and that the MTD was 500 mg. This ratio of 10 between
`the MTD and starting dose was satisfactory in terms of
`safety and efficiency with a total of four dose levels
`investigated in this study.
`
`Critical assessment of the method
`
`The comparative approach is not commonly used and,
`up to the present time, has not been addressed in the
`literature. The differences in the results obtained with the
`different methods would be expected to trigger discussion
`among the scientists
`involved in the starting-dose
`
`841
`
`Table 1. Starting doses for mofarotene as estimated from four
`different methods. A starting dose of 50 mg was selected for the
`‘‘entry into humans’’ (EIH) study in healthy volunteers
`
`Method
`
`Estimated
`dose (mg)
`
`1/3 TDL
`Dose by factor
`Similar drug
`Pharmacokinetically
`guided
`
`57
`23
`250
`277
`
`Safety
`factor
`
`None
`1/10
`1/4
`1/3
`
`Final dose
`(mg)
`
`57
`2.3
`63
`92
`
`estimate regarding the principles and assumptions be-
`hind each approach. This would lead to a better under-
`standing of the science behind the starting-dose estimate.
`Getting similar results with two or three methods such as
`we obtained for mofarotene is reassuring. This method
`could be criticized for being time consuming, but this
`criticism is not considered to be valid by the authors in
`view of the importance of estimating an optimal starting
`dose that is both safe and efficient. It is possible that the
`analysis of different results obtained from various
`methods could be confusing for non-specialists, leading
`to a rather low confidence in the starting-dose estimate.
`However, the alternative approach of relying on a dose
`estimated using a single method also should be ques-
`tioned.
`
`Is the starting dose expected
`to be pharmacodynamically active?
`
`For some compounds, particularly those with a high
`therapeutic index, a starting-dose estimate based on a
`toxicology ‘‘no-effect’’ dose may not be satisfactory.
`This is because, in such compounds, a ‘‘no-effect’’ dose
`in toxicology studies may still have some pharmacody-
`namic activity. In most industrial and academic settings,
`the starting dose is intended to be pharmacodynamically
`inactive, with the second dose having some activity. To
`avoid having a pharmacodynamically active starting
`dose in compounds with a high therapeutic index, the
`starting dose may need to be lower than that estimated
`from toxicology studies.
`The dose that
`is predicted to be the highest
`non-pharmacodynamically active dose in humans can be
`estimated using variations of the dose by factor, similar
`drug, and pharmacokinetically guided approaches that
`utilize data from preclinical pharmacology studies
`instead of toxicology studies. Many variations of the
`dose by factor approach have been described in the lit-
`erature. Posvar and Sedman [6] suggest that the starting
`dose is generally based on a fraction (1/10 to 1/1000) of

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