throbber
DRUG DEVELOPMENT
`
`Selection of the First-Time Dose in Humans:
`
`Comparison of Different Approaches Based
`on Interspecies Scaling of Clearance
`
`Iftekhar Mahmood, PhD, Martin David Green, PhD, and ]. Edward Fisher, PhD
`
`
`
`The authors describefour approaches to selecting a safe start-
`ing dose for humans in clinical drug trials based on
`interspecies scaling of clearance. Human clearance was pre-
`dicted by scaling for 10 example drugs for which animal
`clearance values were available in the literature. The pre-
`dicted human clearance values were then used to select the
`
`estimated starting dose in humans. These doses were then
`compared with the actual doses given to humans during clin-
`ical trials. All four approaches used to estimate the first-time
`dose in humans provided values that were within the dose
`
`range given to humans from Phases I to III. This work demon-
`strates that animal pharmacokinetic data can be used to esti-
`mate a suitable human starting dose, provided the data have
`been obtainedfrom a dose thatproduces no adverse effects.
`
`Keywords: Interspecies scaling; first-time dose; drug devel-
`opment; allometric scaling; predicted human
`clearance; toxicity
`Journal of Clinical Pharmacology, 2003;43:692-697
`@2003 the American College of Clinical Pharmacology
`
`Interspecies scaling is frequently used to predict
`
`pharmacokinetic parameters from animals to hu-
`mans during drug development and is becoming a use-
`ful tool especially for the selection of the first-time dose
`in humans.1 Estimation of a starting dose for “first-in-
`human” clinical trials of new molecular entities in
`
`healthy volunteers is very important since a low start-
`ing dose will prolong dose optimization, and a high
`starting dose may cause serious toxicity. But despite
`the importance of this task from a drug development
`standpoint, there is no consensus regarding the best ap-
`proach for estimating the starting dose.2
`
`From the Division ofClinical Trial Design and Analysis, Centerfor Biologics
`Evaluation and Research (I. Mahmood, M. D. Green) and the Division of
`Neuropharmacological Drug Products, Center for Drug Evaluation and
`Research (J.E. Fisher), Food and Drug Administration (FDA), Rockville,
`Maryland. The views expressed in this article are those of the authors and
`do not reflect the official policy of the FDA. No official support or endorse-
`ment by the FDA is intended or should be inferred. Submitted for publica-
`tion December 22, 2002; revised version accepted April 12, 2003. Ad-
`dress for reprints: lftekhar Mahmood, Division of Clinical Trial Design and
`Analysis, Office of Therapeutic Research and Review, Clinical Pharmacol-
`ogy and Toxicology Branch (HFD-579), Center for Biologics Evaluation
`and Research, Food and Drug Administration, Woodmont Office Center I,
`Suite 200N, 1401 Rockville Pike, Rockville, MD 20852.
`DOI: 10.1177/0091270003254631
`
`One of the most commonly used methods for select-
`ing the starting dose has involved the conversion of an
`animal no observed adverse effect level (NOAEL) to the
`human equivalent dose (HED) using appropriate scal-
`ing factors, followed by application of a safety factor
`(SF). This procedure is described in detail in a recently
`issued Food and Drug Administration (FDA) draft
`guidance, which outlines an algorithmic process for se-
`lecting the maximum recommended starting dose
`(MRSD) for healthy adult volunteers based on animal
`toxicology data and administered doses? Although
`this method has proven generally adequate, it presents
`a number of problems for the toxicologist. Determina-
`tion of the appropriate animal NOAEL is a difficult and
`time-consuming task, depending on a number of key
`study variables, including duration of treatment, dose
`selection, and species. The choice of an appropriate
`scaling factor also involves considerable uncertainty.
`For most systemically administered drugs, conversion
`to the I-IED is typically based on the normalization of
`doses to body surface area.“'5 However, the applicabil-
`ity of the data used to derive this scaling factor (i.e., the
`two-thirds power ofbody weight) to risk assessment for
`noncytotoxic agents and the largely unknown impact
`of experimental conditions on dose scaling across spe-
`
`692 o I Clin Pharmacol 2003;43:692-697
`
`Apotex v. Novartis
`lPR2017-00854
`
`NOVARTIS 2102
`
`Apotex v. Novartis
`IPR2017-00854
`NOVARTIS 2102
`
`

`

`SELECTION OF THE FIRST-TIME DOSE IN HUMANS
`
`cies have been widely discussed.‘*'3 As has been
`pointed out repeatedly, the choice of an SF is almost
`entirely arbitrary and without scientific justification.
`In those cases for which adequate data exist, animal
`pharmacokinetics may aid in determining initial clini-
`cal doses. Although not without their own set of limita-
`tions and assumptions, pharmacokinetically guided
`approaches to dose extrapolation such as those de-
`scribed herein should provide a more rational basis for
`calculating an appropriate starting dose. In 1986, Col-
`lins et al'3 proposed a pharmacokinetically guided dose
`escalation scheme for anticancer drugs. To select the
`first-time dose in humans, Reigner and Blesch2 sug-
`gested the use of the lowest AUC at the NOAEL when a
`drug is given to several species as well as the predicted
`human clearance (dose in humans = AUC X predicted
`human clearance). Therefore, for this approach, it is es-
`sential that the clearance be predicted with reasonable
`accuracy in humans. Over the years, many different ap-
`proaches have been suggested to improve the predic-
`tion of clearance in humans.1°'14 With the introduction
`
`of the rule of exponents by Mahmood and Balian,10 the
`probability of predicting clearance with reasonable ac-
`curacy has vastly improved. The objective of this report
`is to propose four different approaches based on the
`predicted human clearance and their suitability to se-
`lect a safe first-time dose in humans for the Phase I
`
`study.
`
`METHODS
`
`A literature search was conducted to obtain clearance
`
`values for 10 drugs15'33 that have been studied in at least
`three animal species (mice, rat, rabbit, monkey, or dog).
`These drugs are eliminated renally or by extensive me-
`tabolism. Drugs used in this study were given either in-
`travenously or orally. The first step was to predict clear-
`ance in humans. Scaling of clearance was performed
`using the following three methods. Human data were
`not included in the scaling. In the case when several
`doses were given to animals, the lowest dose was se-
`lected for scaling.
`
`Prediction of Clearance
`
`Method I. Clearance of each drug was plotted against
`the body weight on a log-log scale, and the following
`allometric equation was used to predict clearance in
`humans.
`
`CL = a(W)b,
`
`(1)
`
`where W is the body weight and a and b are the coeffi-
`cient and exponent of the allometric equation,
`respectively.
`Method II. The observed clearance values in the dif-
`
`ferent animal species were multiplied by their respec-
`tive maximum life span potential (MLP) and plotted as
`a function of body weight on a log-log scale. From the
`allometric equation, clearance X MLP was estimated in
`humans and the result was then divided by the MLP of
`humans (8.18 x 105 h) to predict the clearance in man.
`
`CL = a(MLP x Clearance)b/8.18 x 105.
`
`(2)
`
`MLP in years was calculated from the following equa-
`tion, as described by Sacher“:
`
`MLP (years) = 135.4 (13W)°-83$ (W)‘°'225,
`
`(3)
`
`where both brain weight (BW) and body weight (W) are
`in kilograms.
`
`Method III. In this approach, clearance of animal
`species was multiplied by the brain weight of the spe-
`cies, and the product was plotted as a function of body
`weight on a log-log scale. The allometric equation
`(equation (4)) was then used to predict the clearance in
`humans using the human brain weight (1.53 kg).
`
`CL = a(CL x BW)b/1.53,
`
`(4)
`
`where BW is the brain weight in kilograms.
`
`Selection of Dose
`
`Based on the predicted human clearance, the following
`four approaches were used to select the dose in hu-
`mans. The predicted dose was then compared with the
`lowest and highest doses given to humans during the
`clinical trials (from Phase I to Phase III).
`
`Approach I. In this approach, the clearances of the
`species (used in the prediction of clearance for hu-
`mans) were plotted on linear scale against the dose
`given to the species. The resultant equation was then
`used to recommend the starting dose in humans as
`follows:
`
`Dose = a + b(x),
`
`(5)
`
`where a is the intercept, b is the slope, and x is the pre-
`dicted clearance in humans.
`
`Approach II. In this approach, the dose given to each
`species was multiplied by the HED and then plotted
`against clearance in each species on a linear scale. The
`resultant equation was then used to select the dose, as
`
`DRUG DEVELOPMENT
`
`693
`
`

`

`MAHZVIOOD ET AL
`
`Table I Names of the Studied Drugs and the Species Used in the Allometric Scaling
`
`
`
` Drugs Species Used Route of Administration“ Reference Animals Reference Humans
`
`
`
`
`
`
`
`17
`15, 16
`Oral
`m, r, (1
`Topiramate
`19
`18
`IV
`m, r, mk, d
`Moxifloxacin
`20
`20
`Oral
`r, d, mk
`Zonisamide
`21
`21
`Oral
`m, r, mk, (1
`Troglitazone
`23
`22
`Oral
`m, r, d, mk
`Venlafaxine
`27
`24-26
`IV
`r, rb, d
`Morphine
`29
`28
`Oral
`r, rb, d
`Felbamate
`30
`30
`Oral
`III, r, mk
`Bepridil
`32
`31
`IV
`m, r, rb, mk
`Stavudine
`
`Zenarestat 33 m, r, d Oral 33
`
`
`
`In, mouse; r, rat; rb, rabbit; mk, monkey; d, dog.
`a. The route of administration in animals and humans was same.
`
`described in equation (5). HED in a given species was
`estimated as followsa:
`
`Dose (mg) = (474 ugOh/mL X 1.26 L/h)/3.27.
`
`HED = animal dose in mg/kg
`>< (animal weight in kg/human weight in kg)°'33.
`
`Approach III. This approach is a slightly modified
`version of the pharmacokinetically guided approach.
`No safety factor was used in this dosing recommenda-
`tion based on the assumption that the dose given to ani-
`mals is much lower than the NOAEL. Since the AUC
`
`values for a given drug were available in more than one
`species, the recommended dose was estimated using
`the lowest AUC observed in a given species as follows:
`
`Dose (mg) = AUC in animal (pg-h/mL)
`>< predicted clearance in humans (L/h).
`
`Approach IV This mathematically manipulated ap-
`proach can be used to recommend the first-time dose in
`humans as follows.
`
`First, one selects the species whose clearance (per kg
`body weight) is nearest to the predicted human clear-
`ance (based on kg body weight). A correction factor is
`then obtained by dividing the clearance of the chosen
`species by the predicted human clearance. Then the
`recommended dose can be selected according to the
`following equation:
`
`Dose (mg) = (AUC in the chosen animal (ugOh/mL)
`X predicted clearance in humans (L/h))/correction factor.
`
`Example: For felbamate, the observed clearance in rat,
`rabbit, and dog was 2.54, 0.98, and 1.55 mL/min/kg, re-
`spectively. In man, the predicted clearance was 0.3 mL/
`min/kg. Based on the clearance value, rabbit was the
`closest species to man. The estimated correction factor
`was 3.27 (098/03), and the dose was calculated as
`follows:
`
`694 o I Clin Pharmacol 2003;43:692-697
`
`Thus, the recommended starting dose of felbamate in
`humans was 183 mg.
`
`RESULTS
`
`Table I is the summary of the drugs and species used in
`this study as well as the route of administration of these
`drugs to animals and humans. The exponents of the
`simple allometry and the correlation coefficient be-
`tween body weight and clearance are summarized in
`Table II. Table II also compares the predicted and ob-
`served clearance of the studied drugs.
`A good correlation between body weight and clear-
`ance was observed for most of the drugs tested. The re-
`sults of this study (as in the previous study”) indicate
`that there are specific conditions under which only one
`of the three methods can be used for improved predic-
`tion of clearance. When the exponents of the simple
`allometry ranged from 0.55 to 0.70, the simple
`allometry was considered suitable for the prediction of
`clearance in humans. When the exponents of the sim-
`ple allometry ranged from 0.70 to 0.99, the MLP ap-
`proach was found to be appropriate for the prediction
`of clearance in humans. The product of clearance and
`brain weight was considered suitable for the prediction
`of clearance in humans when the exponents of the sim-
`ple allometry were 2 1.0. It can be seen fiom Table II
`that simple allometry was not adequate for the predic-
`tion of clearance for all drugs, but the use of the rule of
`exponents vastly improved the prediction of these
`drugs.
`In this study, however, there were two drugs whose
`predicted clearances were many times higher than the
`observed clearances. Zenarestat’s exponent was 1.340,
`but as mentioned previously,10 if the exponent of the
`
`

`

`SELECTION OF THE FIRST-TIME DOSE IN HUMANS
`
`Table II Observed versus Predicted Clearance (mL/min) in Humans
`
`
`Drugs
`Exponent
`Correlation Coefficient (1')
`Observed CL
`Predicted CL (SA)
`Predicted CL (RE)
`
`Topiramate
`Moxifloxacin
`Zonisamide
`
`Troglitazone
`Venlafaxine
`
`Morphine
`Felbamate
`
`0.557
`0.603
`0.735
`
`0.633
`0.782
`
`0.777
`0.823
`
`0.864
`0.969
`0.987
`
`0.984
`0.866
`
`0.985
`0.979
`
`22-36
`154
`16
`
`751-891
`2240
`
`1300
`30
`
`51
`166
`22
`
`793
`4874
`
`2714
`59
`
`51
`166
`20
`
`793
`2268
`
`910
`21
`
`6535
`12,886
`1085
`0.995
`0.842
`Bepridil
`Stavudine
`0.901
`0.999
`572
`961
`400
`
` Zenarestat 1.340 0.994 47 688 155
`
`
`
`
`
`
`SA, simple allometry; RE, rule of exponents.
`
`allometry is > 1.30, the predicted clearance may be
`many times higher than the observed clearance, and in-
`deed this was the case with zenarestat. In a previous pa-
`per,35 Mahmood mentioned that the prediction of oral
`clearance would be erratic when the oral clearance of
`
`the species used in the scaling is either equal to or
`greater than the liver blood flow and the observed hu-
`man clearance is less than the liver blood flow. This ob-
`
`servation was found to be true with bepridil. Bepridil’s
`oral clearance in the mouse, rat, and monkey was
`greater than their respective blood flow, resulting in a
`much higher prediction of oral clearance (observed hu-
`man CL = 1085 mL/min, which is less than human liver
`
`blood flow) in humans (predicted CL = 6535 mL/min).
`The results obtained by the four approaches to se-
`lecting the first-time dose in humans are compared
`with the doses actually given to humans from Phase I to
`Phase III in Table III. The clinical dose range shown ex-
`tends from the first human dose to the highest dose
`used in the definitive efficacy trials, which can be con-
`sidered a well-tolerated but not necessarily a maximum-
`tolerated dose. The method of dose selection in the
`
`original trials is unknown. Approach I was the most ag-
`gressive approach and may not be suitable for the selec-
`tion of the first-time dose in humans. Approach III was
`the most conservative method and, like Approach I,
`may be unsuitable. Approaches II and IV were more
`moderate and could be useful in estimating a safe and
`efficient dose for the first-time administration of a drug
`to humans. It should be noted, however, that with the
`
`exception of zenarestat, even the dose arrived at by Ap-
`proach I did not exceed the highest dose given to hu-
`mans for any of the studied drugs.
`
`DISCUSSION
`
`Interspecies scaling is based on the assumption (a cor-
`rect assumption) that there are anatomical, physiologi-
`cal, and biochemical similarities among animals,
`which can be described by mathematical models. It is
`now a well-established fact that many physiological
`processes and organ sizes exhibit a power-law relation-
`ship with the body weight of species. This relationship
`is the scientific basis of allometry.
`Pharmacokinetics play an important role during
`drug development. Characterization of absorption,
`distribution, metabolism, and excretion (ADME) in
`animals is of fundamental importance. The pharma-
`cokinetic parameters, especially clearance, can be ex-
`trapolated to humans for the selection of a safe dose for
`the first-dosing trials in humans. Two of the widely
`used methods of first-time dose selection are based on
`
`the NOAEL or toxicokinetic studies of a drug in one to
`three species. There are several disadvantages to these
`approaches. In the NOAEL approach, it becomes nec-
`essary to select a NOAEL, which is a tedious and time-
`consuming process. In reality, one may never find an
`absolute NOAEL in a given species. In toxicokinetic
`studies, animals are given a very high dose chronically,
`and if the resultant toxicity does not kill the animal, it
`may alter the physiology of the animal. This change in
`the physiology of the animal may have an impact on
`the pharmacokinetics of a given drug. Overall, both the
`NOAEL and toxicokinetic approaches for the first-time
`selection of a dose to humans are slow and very con-
`servative approaches. In fact, there is no need to select
`the first dose in humans based on NOAEL or
`
`DRUG DEVELOPMENT
`
`695
`
`

`

`MAHIVIOOD ET AL
`
`Table III Recommended First-Time Dose in Humans by Different Approaches
`
`
`
` Drugs Approach I Approach II Approach III Approach IV Dose Given
`
`
`
`
`
`
`
`
`
`Topiramate
`Moxifloxacin
`Zonisamide
`
`Troglitazone
`Venlafaxine
`
`Morphine
`Felbamate
`
`175
`107
`595
`
`90
`60
`
`9.3
`145
`
`95
`86
`329
`
`37
`23
`
`5.3
`80
`
`36
`23
`265
`
`35
`22
`
`2.5
`122
`
`44
`80
`490
`
`85
`60
`
`3
`183
`
`100-1200
`84-400
`200-800
`
`100-600
`25-150
`
`7.5-10
`100-800
`
`200-400
`176
`161
`177
`400
`Bepl‘idil
`47-280a
`134
`170
`86
`243
`Stavudine
`
`
`
`
`
`Zenarestat 150-600 845 460 313 353
`
`Approach I = dose versus clearance (CL); Approach II = human equivalent dose (HED) dose versus CL; Approach III = no observed adverse effect level (NOAEL);
`Approach IV = correction factor.
`a. Given orally, but the recommended dose is based on the scaling of IV data as the absolute bioavailability is 1 in humans. The initial IV dose in humans was
`70 mg.
`
`toxicokinetics. One can achieve this goal by giving a
`safe low dose to animals and, based on the prediction of
`clearance, selecting a safe and suitable dose in humans
`as described in this study. Nevertheless, this does not
`necessarily mean that one should not try to determine
`the NOAEL or conduct a toxicokinetic study as these
`endpoints provide information that is often useful in
`characterizing toxicity.
`In this report, we have proposed several methods
`that can be used to administer a first-time dose to hu-
`
`mans that is not only safe but also avoids many unnec-
`essary low-dose schemes. Even when the predicted
`clearance is many times higher than the observed clear-
`ance, as seen with bepridil (sixfold higher) and
`zenarestat (threefold higher), the doses selected by Ap-
`proaches II, III, and IV were well below the highest
`well-tolerated doses given to humans. It is, however,
`not known that these clinical doses represent the maxi-
`mum tolerated dose or whether one could give even
`higher doses to humans without producing any signifi-
`cant toxicity. Approach III is, in fact, a modified version
`of NOAEL or toxicokinetic methods for the selection of
`
`the first-time dose in humans. In this approach, a safety
`factor was not used since the animal doses were low
`
`and without any side effects.
`An important caveat is that the above-mentioned ap-
`proaches ignore the fact that there may be a toxic me-
`tabolite of a drug in humans that is not formed in ani-
`mals. Considering that current standards of drug
`development require a thorough investigation of me-
`tabolites formed in animals and humans, such a case is
`highly unlikely to occur.2 Another important point
`worth considering is that genetic polymorphism, as in
`the case of 2D6, may occur. One may then require de-
`
`veloping two different starting doses, one for poor
`metabolizers and one for extensive metabolizers.z
`
`The main objective of this work is to describe differ-
`ent approaches that can help to select a first dose in hu-
`mans that is not only safe but also efficient (neither very
`low nor very high). These suggested approaches pro-
`vide rational alternatives to the somewhat arbitrary
`dose selection process often used. Having a clear set of
`methods rather than relying on some fuzzy approach
`should be an important advantage in the bigger context
`of drug development. Although various safety factors
`used in the first-time dose in humans have no scientific
`basis, one can tailor these factors based on the need or
`
`the characteristics of a given drug. It should also be
`noted that there is no right or wrong method, and one
`can select one of the many approaches available by us-
`ing scientific judgment and the ease of the method. Our
`proposed methods are one of the many approaches that
`may be used to select a first-time dose to humans. Other
`approaches may be considered in relationship to the
`severity and incidence of toxicity. For example, a con-
`servative approach is suitable for drugs likely to have a
`narrow therapeutic index.
`
`REFERENCES
`
`1. Boxenbaum H, Dilea C: First-time-in—human dose selection:
`allometric thoughts and perspectives. I Clin Pharmacol 1995;35:957-
`966.
`
`2. Reigner BG, Blesch KS: Estimating the starting dose for entry into
`humans: principles and practice. Eur] CIin Pharmacol 2002;57:835-
`845.
`
`3. US. Food and Drug Administration: Draft guidance for industry
`and reviewers: estimating the safe starting dose in clinical trials for
`
`696 o I Clin Pharmacol 2003;43:692-697
`
`

`

`SELECTION OF THE FIRST-TIME DOSE IN HUMANS
`
`therapeutics in adult healthy volunteers. Fed Reg 2003;68:2340-
`2341.
`
`4. Freireich EI, Gehan EA, Rall DP, Schmidt LH, Skipper HE: Quanti-
`tative comparison of toxicity of anticancer agents in mouse, rat, ham-
`ster, dog, monkey, and man. Cancer Chemother Rep 1966;50:219-
`244.
`
`5. Shein PS, Davis RD, Carter S, Newman I, Shein DR, Rall DP: The
`evaluation of anticancer drugs in dogs and monkeys for the predic-
`tion of qualitative toxicities in man. Clin Pharmacol Therap 1979;
`11:3-40.
`
`6. Travis CC, Bowers IC: Interspecies scaling of anesthetic potency.
`Toxicol Indust Health 1991;7:249-260.
`
`7. Watanabe K, Bois FY, Zeise L: Interspecies extrapolation: areexam-
`ination of acute toxicity data. Risk Anal 1992 ;2:301-310.
`8. Rhomberg LR, Wolff SK: Empirical scaling of single oral lethal
`doses across mammalian species based on a large database. HiskAnal
`1998;18:741-753.
`
`9. Collins IM, Zaharko S, Dedrick RL, Chabner BA: Potential roles for
`preclinical pharmacology in phase I clinical trials. Cancer Treat Rep
`1986;70:73-80.
`
`10. Mahmood I, Balian ID: Interspecies scaling: predicting clearance
`of drugs in humans. Three different approaches. Xenobiotica
`1996;26:887-895.
`
`11. Boxenbaum H, Fertig IB: Scaling of antipyrine intrinsic clearance
`of unbound drug in 15 mammalian species. Eur I Drug Metab
`Pharmacokin 1984;92177-183.
`
`12. Lave TH, Dupin S, Schmitt C, Chou RC, Iaeck D, Coassolo PH: In-
`tegration of in vitro data into allometric scaling to predict hepatic
`metabolic clearance in man: application to 10 extensively metabo-
`lized drugs. IPharm Sci 1997;86:584-590.
`13. Obach RS, Baxter IG, Liston TE, Silber BM, Iones C, Macintyre F,
`et al: The prediction of human pharmacokinetic parameters from
`pre-clinical and in vitro metabolism. I Pharmacol Exp Ther 1997;
`283:46-58.
`
`14. Boxenbaum H: Interspecies pharmacokinetic scaling and the
`evolutionary-comparative paradigm. Drug Metab Rev 1984;15:1071-
`1 1 2 1.
`
`15. Streeter AI, Stahle PL, Holland ML, Pritchard IF, Takacs AR:
`Pharmacokinetics and bioavailability oftopiramate in the beagle dog.
`Drug Metab Dispos 1995;23:90-93.
`16. Streeter AI, Stahle PL, Hills IF, Lucas PN, McKown LA, Nese SL,
`et al: Pharmacokinetics of topiramate in the rat. Pharm Res 1994;
`11(Suppl.):SS72.
`17. Rosenfeld WE: Topiramate: a review of preclinical, pharma-
`cokinetic, and clinical data. Clin Ther 1997;19:1294-1308.
`
`18. Siefert HM, Domdey—Bette A, Henninger K, Hucke F, Kohlsdorfer,
`Stass HH: Pharmacokinetics of the 8-methoxyquinolone,
`moxifloxacin: a comparison in humans and other mammalian spe-
`cies. I Antimicrob Chemotherap 1999;43(Suppl. B):69-76.
`19. Stass H, Kubitza D: Pharmacokinetics and elimination of
`moxifloxacin after oral and intravenous administration in man. I
`Antimicrob Chemotherap 1999;43(Suppl. B):83-90.
`20. Matsumoto K, Miyazaki H, Fuji T, Maeda T, Hashimoto M: Ab-
`sorption, distribution and excretion of 3-(sulfanoyl [14C] methyl)-
`
`1,2-benzisoxazole—(AD-810) in rats, dogs and monkeys and of AD-
`810 in men. Arzneim-Forsch 1983;33:961-967.
`21. Izami T, Enomoto S, Hosiyama K, Sasahara K, Shibukawa A,
`Nakagawa T, et al: Pharmacokinetics of troglitazone, a new and ex-
`tensively metabolized antidiabetic agent, after oral administration,
`with an animal scale-up approach. I Pharmacol Exp Ther 1996;
`277:1630-1641.
`
`22. Howell SR, Hicks DR, Scatina IA, Sisenwine SF: Pharma-
`cokinetics of venlafaxine and o-desmethylvenlafaxine in laboratory
`animals. Xenobiotica 1994;24:315-327.
`
`23. Klamerus KI, Maloney K, Rudolph RL, Sisenwine SF, Iusko WI,
`Chiang ST: Introduction of a composite parameter to the pharma-
`cokinetics of venlafaxine and its active o-desmethyl metabolite. I
`Clin Pharmaco]1992;32:716-724.
`
`24. Bhargava HN, Villar VM, Gulati A, Chari G: Analgesic and
`hyperthermic effects of intravenously administered morphine in the
`rat are related to its serum levels. I Pharmacol Exp Therap 1991;
`258:511-516.
`
`25. Matsuzawa T, Wada Y, Shimoyama M, Nakajima K, Seki T,
`Sugibayashi K, et al: The effect of different routes of administration
`on the metabolism of morphine: the disposition of morphine and its
`metabolites after topical application. Biopharm Drug Disp 1994;
`15:665-678.
`
`26. Merrell WI, Gordon L, Wood AI, Shay S, Iackson EK, Wood M:
`The effect of halothane on morphine disposition: relative contribu-
`tions of the liver and kidney to morphine glucuronidation in the dog.
`Anesthesiology 1990;72:308-314.
`27. Miline RW, Nation RL, Somogyi AA: The disposition of morphine
`and its 3-and 6-glucuronide metabolites in humans and animals and
`the importance of the metabolites to the pharmacological effects of
`morphine. Drug Metab Rev 1996;28:345-472.
`28. Adusumalli VE, Yang IT, Wong KK, Kucharczyk N, Sofia RD:
`Felbamate pharmacokinetics in the rat, rabbit, and dog. Drug Metab
`Disp 1991;19:1116-1125.
`29. Physician’s Desk Reference. 56th ed. Montvale, NI: Medical Eco-
`nomics Company, 2002.
`30. Wu WN, Pritchard IF, Hills IF, Uetz IA, Yorgey KA, McKown LA,
`et al: Disposition of bepridil in laboratory animals and man.
`Xenobiotica 1992;22:153-169.
`
`31. Kaul S, Dandekar KA, Schilling BE, Barbhaya RH: Toxicokinetics
`of 2’, 3’-didehydro-3’-deoxythymidine, stavudine (D4T). DrugMetab
`Disp 1999;27:1-12.
`32. Rana KZ, Dudley MN: Clinical pharmacokinetics of stavudine.
`Clin Pharmacokin 1997;33:276-284.
`
`33. Tanaka Y, Shimojyo H, Hata T, Hashimoto M, Noguchi H:
`Toxicokinetics of zenarestat, an aldose reductase inhibitor in animals
`and man. Xenobiotica 1994;24:461-471.
`
`34. Sacher G: Relation of lifespan to brain weight and body weight in
`mammals, in: Wolstenholme GEW, O’Connor M (eds.), CIBA Foun-
`dation Colloquia on Aging. London: Churchill, 1959;115-133.
`35. Mahmood I: Interspecies scaling: predicting oral clearance in hu-
`mans. AmericanITherap 2002;9z35-42.
`
`DRUG DEVELOPMENT
`
`697
`
`

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