throbber
-,-, 1/i
`
`1 r ~r) ·1
`
`! . ~
`
`_ I
`
`Integration of
`Pharmacokinetics,
`Pharmacodynamics,
`and Toxicokinetics
`in Rational
`Drug Development
`
`Apotex v. Novartis
`IPR2017-00854
`NOVARTIS 2071
`
`1
`
`

`

`Integration of
`Pharmacokinetics,
`Pharmacodynamics,
`and Toxicokinetics
`in Rational
`Drug Development
`
`2
`
`

`

`Integration of
`Pharmacokinetics,
`Pharmacodynamics,
`and Toxicokinetics
`in Rational
`Drug Development
`
`Edited by
`
`Avraham Yacobi
`
`American Cyanamid Company
`Pearl River, New York
`
`Jerome P. Skelly
`
`Food and Drug Administration
`Washington, D.C.
`
`Vi nod P. Shah
`
`Food and Drug Administration
`Rockville, Maryland
`and
`Leslie Z. Benet
`
`University of California, San Francisco
`San Francisco, California
`
`.
`.
`Published in cooperation with the
`American Association of Pharmaceutical Sc1ent1sts
`
`Plenum Press• New York and London
`
`3
`
`

`

`Library of Congress Catalog1ng-1n-Publ1cat1on Data
`
`Integration of pharmacokinetics, pharmacodynamics, and toxicokinetics
`in rational drug development / edited by Avraham Yacobi ... let
`a 1. ].
`cm.
`p.
`"Sponsored by the American Association of Pharmaceutical
`Scientists, the U.S. Food and Drug Administration, and the American
`Society for Cl lnical Pharmacology and Therapeutlcs"--T.p. verso.
`"Published In cooperation with the American Association of
`Pharmaceutical Scientists."
`Includes bibliographical references and Indexes.
`ISBN 0-306-44356-2
`1. Drugs--Research--Methodology--Congresses. 2. Pharmacoklnetlcs(cid:173)
`-Congresses. 3. Drugs--Physiologica I effect--Congresses. 4. Drugs-
`-Toxicology--Congresses.
`I. Yacobi, Avraham.
`II. American
`Association of Pharmaceutical Scientists.
`III. United States. Food
`and Drug Administration.
`IV. American Society for Clinical
`Pharmacology and Therapeutics.
`[DNLM: 1. Drug Evaluation--congresses. 2. Drug Screening(cid:173)
`-congresses. 3. Drugs, Investigatlonal--congresses.
`av 771 I602l
`4. Pharmacoklnetics--congresses.
`RM301.25.I58 1993
`615' .1 '072--dc20
`DNLM/DLC
`for Library of Congress
`
`92-48285
`CIP
`
`Invited papers presented at a conference entitled "The Integration of Pharmacokinetic,
`Pharmacodynamic, and Toxicokinetic Principles in Rational Drug Development," held
`April 24-26, 1991, in Arlington, Virginia, sponsored by the American Association of
`Pharmaceutical Scientists, the U.S. Food and Drug Administration, and the American
`Society for Clinical Pharmacology and Therapeutics
`
`ISBN 0-306-44356-2
`
`© 1993 Plenum Press, New York
`A Division of Plenum Publishing Corporation
`233 Spring Street, New York, N.Y. 10013
`
`All rights reserved
`
`No part of this book may be reproduced, stored in a retrieval system, or transmitted
`in any form or by any means, electronic, mechanical, photocopying, microfilming,
`recording, or otherwise, without written permission from the Publisher
`
`Printed in the United States of America
`
`4
`
`

`

`CONTENTS
`
`INTRODUCTION
`
`SECTION
`I
`
`INTRODUCTION
`
`1. Rationale for the Effective Use of Pharmacokinetics and Pharmacodynamics in
`Early Drug Development
`Carl C. Peck
`
`12. The Case for Preclinical Pharmacodynamics
`
`Gerhard Levy
`
`xv
`
`l
`
`7
`
`3. Utility of Kinetic, Dynamic, and Metabolic Data in Nonclinical
`Pharmacology{foxicology Studies
`
`Judi Weissinger
`
`15
`
`SECTION
`II
`
`PRECLINICAL PHARMACOKINETIC & PHARMACODYNAMIC
`STUDIES
`· 4. Pharmacokinetics and Drug Metabolism in Animal Studies (ADME, Protein
`Glenna G. Fitzgerald
`Binding, Mass Balance, Animal Models)
`! 5. Nonclinical Considerations: Disposition of Drugs
`
`Martin David Green
`
`"6. Use of Acute Toxicity Data in the Design and Interpretation of Subchronic and
`Chronic Toxicity Studies
`Karl K. Rozman
`
`I
`
`17. Use of Pharmacokinetics and Pharmacodynamics in Preclinical Studies to Guide
`Dosage Escalation Schemes in Phase I Studies of Anticancer Drugs
`Jerry M. Collins
`
`J 8. Use of Toxicokinetic Principles in Drug Development: Bridging Preclinical and
`Mauricio Leal, Avraham Yacobi and Vijay K. Batra
`Clinical Studies
`
`23
`
`33
`
`39
`
`49
`
`55
`
`xi
`
`5
`
`

`

`SECTION
`III
`
`METHODOLOGY FOR PRECLINICAL PHARMACODYNAMIC STUDIES
`
`9. Preclinical Phannacodynamics of Central Nervous System Active Agents
`Meindert Danlzof, Jaap W. Mandema and Arendien Hoogerkamp
`
`10. Preclinical Pharmacodynamics of Antihypertensives
`Robert A. Branch and Edwin K. Jackson
`
`11. Preclinical Pharmacodynamics of Anxiolytics: Effects of Chronic
`Benzodiazepine Administration
`Lawrence G. Miller
`
`69
`
`81
`
`91
`97
`
`12. Guidelines for Development of a New Diuretic Agent
`13. Preclinical Pharmacodynamics of Anti-Inflammatory Drugs
`Asoke Mukherjee, Conrad Chen, Lucy Jean and Claude B. Coutinho 105
`
`D. Craig Brater
`
`SECTION
`IV
`
`PRECLINICAL & CLINICAL PIIARMACOKINETICS
`
`J 14. The Role of Pharmacokinetics in the Drug Development Process
`Leslie z. Benet 115
`15. Implementation of an Effective Pharmacokinetics Research Program in Industry
`Avraham Yacobi, Vijay K. Batra, Robert Desjardins, Robert D. Faulkner,
`Gabriela Nicolau, William R. Pool, Anita Shah and Alfred R Tonelli 125
`
`16. Pharmacoepidemiology, Population Pharmacokinetics and New
`Thaddeus fl. Grasela and Edward J. Antal 137
`Drug Development
`
`17. Assessment of Pharmacokinetic Drug Interactions in Clinical Drug Development
`Jerome J. Schentag 149
`
`SECTION
`V
`
`CLINICAL PHARMACODYNAMICS
`
`18. Pharmacokinetic/Pharmacodynamic Models and Methods
`Davide Verotta and Lewis B. Sheiner 159
`
`19. Pharmacokinetic and Pharmacodynamic Modeling Applied to Anesthetic Drugs
`Donald R Stanski 179
`
`20. Pharmacodynamic/Pharmacokinetic Relationships for Rapidly Acting Drugs
`(NSAIDS) in Rheumatoid Arthritis: Problems and Preliminary Solutions
`Daniel E. Furst 193
`
`xii
`
`This material was c,opied
`
`6
`
`

`

`21. The Value of Plasma-Warfarin Measurement
`
`22. Clinical Phannacodynamics of Cardiovascular Agents:
`Focus on Sudden Cardiac Death
`
`23. The Benzodiazepines: Kinetic-Dynamic Relationships
`
`RobertA.O'Reilly 201
`
`Dan M. Roden 207
`
`David J. Greenblatt 217
`
`SECTION
`VI
`
`APPLICATION OF PIIARMACODYNAMICS & PHARMACOKINETICS IN
`THE DRUG DEVELOPMENT PROCESS
`
`24. The Integration of Phannacodynamics and Pharmacokinetics in Rational Drug
`Development
`Sally Usdin Yasuda, Sorell L. Schwartz, Allton Wellstein and
`Raymond L. Woosley
`
`25. Concentration-Controlled Trials: Basic Concepts, Design, and Implementation
`
`225
`
`lilly Sanathanan and Carl C. Peck 239
`
`SECTION
`VII
`
`SUMMARY REPORT
`
`26. Opportunities for Integration of Pharma co kinetics, Pharmacodynamics and
`Toxicokinetics in Rational Drug Development
`
`A~m~~
`
`Subject Index
`
`249
`
`~
`
`267
`
`This material was<:opied
`at th,e NLMand may he
`
`xiii
`
`7
`
`

`

`THE CASE FOR PRECLINICAL PHARMACODYNAMICS
`
`Gerhard Levy
`Department of Pharmaceutics, School of Pharmacy
`State University of New York at Buffalo, Amherst, N.Y. 14260
`
`ABSTRACT
`
`Pharmacodynamics concerns the relationship between drug concentration and intensity
`of pharmacologic effect. It includes the exploration and assessment of relevant variables such
`as pharmacologically active metabolites, pharmacodynamic drug interactions and effects of
`genetics and underlying diseases. Available (though limited) infonnation indicates that drug
`concentrations required to produce a defined intensity of pharmacologic action are quite
`similar in man and animals. For such interspecies comparisons, consideration must be given
`to possible differences in drug-protein binding, active metabolites, kinetics of equilibration
`of drug between plasma and biophase, and definition of the pharmacologic endpoint. It is now
`evident that many unanticipated, phannacodynamically relevant clinical risk factors, includ(cid:173)
`ing some that have caused a substantial number of deaths, could have been recognized in
`preclinical pharmacodynamic studies. Examples based on studies in the author's laboratory
`include potentiation of barbiturates in renal failure, increased response to benzodiazepines in
`severe liver disease, increased neurotoxicity of theophylline in renal failure, increased potency
`of general anesthetics in hemorrhagic hypovolemia, and the steep responder syndrome in
`warfarin therapy. Preclinical pharmacodynamics provides a means for identifying, in advance
`of clinical use, potential risk factors and conditions under which drugs may be subject to
`profound changes in their concentration-effect relationships.
`
`INTRODUCTION
`
`If the process of drug development is to be productive and efficient, the various phases
`of this process must be designed to elicit information which is useful for the subsequent phases
`of development. There must be a logical continuum wherein previous infonnation is used as
`a basis for the design of the next studies. This is particularly important in the planning of
`
`!111egra1io11 of Pharmacokinelics, Pharmacodynamics, and Toxicoki11etics in Ralional
`Drug Developme11/, Edited by A. Yacobi el al., Plenum Press, New York, 1993
`
`7
`
`8
`
`

`

`clinical investigations and the development of clinical protocols because inadequate clinical
`studies can contribute substantially to the cost of a drug development and to the time required
`to bring a new drug on the market.
`
`PRECLINICAL PHARMACOKINETICS AND PHARMACODYNAMICS
`
`The place of preclinical (animal) phannacokinetics in drug development is well estab(cid:173)
`lished. Assessment of drug absorption and elimination kinetics is done as part of the selection
`of the most promising in a series of pharmacologically active compounds. Studies in animals
`are performed also to determine if the candidate compounds are enzyme inducers or inhibitors,
`or if they are subject to other time-dependent effects such as product inhibition or cosubstrate
`depletion. If such is the case, then future clinical studies must be designed to assess the
`magnitude and time course of these alterations in the therapeutically relevant drug concentra(cid:173)
`tion range. Determination of the pharmacokinetics of the drug candidate in several animal
`species is the logical precursor of well-designed animal toxicology studies and provides the
`opportunity to explore interspecies correlations with the ultimate aim of predicting, by
`appropriate extrapolation, the pharmacokinetic characteristics of the compound in man.
`Unlike pharmacokinetics, the role of pharmacodynamics in the preclinical phase of drug
`development is barely in its beginning. This is unfortunate because pharmacodynamics, i.e.
`the relationship between drug concentration in plasma or certain tissue fluids and the intensity
`of drug action, is essential to the optimum interpretation and utilization of phannacokinetic
`information. At present, only one relatively minor aspect of preclinical pharmacodynamics
`appears to be an established part of the drug development process. That aspect is the
`assessment of the pharmacologic activity of the metabolites of a drug or drug candidate. The
`relevance of this assessment to the drug discovery process is obvious. There are, however,
`other aspects of preclinical pharmacodynamics that are important and yet frequently ne(cid:173)
`glected. These include in the first instance the characterization of the relationship between
`drug concentration and intensity of drug action(s), preferably in sufficient detail to permit
`modelling and to recognize time-dependent phenomena such as the development of functional
`tolerance. Several other aspects of preclinical pharmacodynamics can contribute substantially
`to the drug development process. One of these is the use of animal data to predict therapeutic
`and toxic drug concentrations in humans.
`
`EFFECTIVE DRUG CONCENTRATIONS IN ANIMALS AND HUMANS
`
`The plasma concentration of a drug required to elicit a certain (intensity of) action is
`often similar in experimental animals and humans. In a report on the Workshop on the
`Qualitative and Quantitative Comparability of Human and Animal Developmental Neuro(cid:173)
`toxicity, Francis, Kimmel, and Rees (1990) stated that "Comparisons of administered doses
`(resulting in similar effect levels) revealed a wide range of differences across species (up to
`a 10,000-fold difference). On the other hand, comparisons across species using internal
`
`8
`
`9
`
`

`

`(
`t'
`measurements of dose (e.g., blood or brain levels) showed a rema k bl
`
`.
`,,
`11 r a e corre a 1011 genera y,
`1
`a 1 to 2-fold difference). In the case of the antiepileptic drug phenytoin, Ramzan (1990)
`reported that the onset of ataxia occurred in rats at serum unbound d
`t
`t'
`f 6 3
`rug concen ra ions o
`.
`± 1.7 (mean± S.D.) mg/1. The serum total phenytoin concentrations associated with ataxia in
`humans are between 30 and 40 mg/1 (Kutt and co-workers, 1964); the corresponding
`concentrations of unbound drug ar~ between 3 and 6 mg/I based 011 the typical phenytoin
`protein binding of 10 to 15 percent 111 human serum. In another study, neurotoxic manifesta(cid:173)
`tions of phenytoin in pregnant rhesus macaques occurred at plasma concentrations (total drng)
`in excess of 30 µg/ml, "consistent with data reported in mouse, rat, rabbit, dog, rhesus macaque
`and human" (Hendrie and co-workers, 1990). The plasma concentration of the antihyperten(cid:173)
`sive agent nilvadipine associated with a 20 percent reduction of diastolic blood pressure was
`about 20 mg/ml in dogs (Wu and co-workers, 1988) and the same in humans (Cheung and
`co-workers, 1988). The concentration of ethanol in brain, serum and cerebrospinal fluid of
`rats at onset of loss of righting reflex (i.e. sleep) is between 3 and 4 mg/ml or mg/g (Danhof,
`Hisaoka and Levy, 1985); the hypnotic concentrations of ethanol in man are similar.
`It is unfortunate that comparative information on the effective concentrations of new
`drugs in various species of animals and ( eventually) humans is either not obtained or not fully
`utilized in drug development. Knowledge of effective drug concentrations in animals can
`greatly facilitate dose ranging in Phase I studies. There is increasing evidence that, as Gianni
`and co-workers ( 1990) have stated, "preclinical phannacology offers reliable guidelines to
`safely accelerate the process of Phase I trials".
`One must, however, be aware of several important considerations when making inter(cid:173)
`species comparisons of effective drug concentrations. First, the phannacologic endpoint used
`for detenninations of effective concentration must be comparable in the different species.
`Next, attention must be paid to the appropriate fluid or tissue for concentration determination
`in the context of the possibility of a disequilibrium between drug in plasma and drng at the
`site of action (Danhof and Levy, 1984). Differences in plasma protein binding of a drug
`between species must be taken into consideration; concentrations of unbound drug should be
`compared in cases where drug-protein binding is extensive. Also, the possible contribution of
`active metabolites to the pharmacologic effect of a drug must be kept in mind.
`
`ASSESSMENT OF COMPLEX PHARMACODYNAMIC RELATIONSHIPS
`
`Studies in experimental animals can reveal and elucidate complex pharmacodynamic
`relationships which also occur in humans. An excellent example are the pharmacodynamics
`of the coumarin anticoagulants. These agents, of which warfarin and dicumarol are best known
`in the United States, act indirectly and their maximum clinically apparent effect (increase of
`prothrombin time) occurs in man about 2 days after maximum plasma concentrations
`produced by a single, large dose (Nagashima, O'Reilly and Levy, 1969). We have developed
`a pharmacodynamic model which permits resolution of the observed change in prothrombin
`complex activity (which is derived from the prothrombin time) into its components: the
`synthesis and elimination of prothrombin complex activity. The direct effect of the coumarin
`
`9
`
`10
`
`

`

`anticoagulants is the inhibition of the synthesis of prothrombin complex activity (which
`reflects the inhibition of the synthesis of vitamin K dependent clotting factors) and this
`inhibitory effect is related linearly to the logarithm of the drug concentration in plasma without
`any delay (Nagashima, O'Reilly and Levy, 1969). The pharmacodynamic model includes the
`assumption that prothrombin complex activity declines exponentially with time after admini(cid:173)
`stration of a synthesis-blocking dose of the coumarin anticoagulant. Despite the complexity
`of the pharmacodynamic model, it was found to apply equally to humans and rats (Yacobi,
`Wingard and Levy, 1974). This exemplifies the possibility of exploring pharmacodynamic
`modelling approaches in preclinical studies for the purpose of optimizing subsequent clinical
`protocols.
`Another of our studies of warfarin phannacodynamics in rats provided additional,
`valuable information relevant to the therapeutic use of the drug in humans. Warfarin pharma(cid:173)
`codynamics, like the pharmacodynamics of other drugs (Levy, 1985), exhibit pronounced
`inter-individual but relatively little intraindividual variability in humans. We observed such
`inter-individual variability also in rats and found a strong correlation between the slope of the
`anticoagulant effect-log concentration relationship of warfarin and the free fraction value of
`the drug in the serum (Yacobi and Levy, 1977). Based on that observation, Routledge and
`co-workers (1979) recognized a similar relationship in man. Since the serum or plasma free
`fraction value of warfarin can be determined by the in vitro addition of the drug to a drug-free
`serum or plasma sample, one is able to characterize "steep" and "shallow" warfarin responders
`before initiation of therapy.
`
`EFFECT OF DISEASE STATES ON THE KINETICS OF DRUG ACTION
`
`There are many reasons for the large interindividual variability in the pharmacody(cid:173)
`namics of drugs (Levy, 1985). One such reason is the presence of underlying disease. Our
`group has intensively explored this aspect of pharmacodynamics in recent years. One of our
`first efforts in this area was stimulated by a report in 1954 by Dundee and Richards of
`significantly lower loading and maintenance dose requirements for thiopental-mediated
`general anesthesia in uremic patients as compared to patients with normal renal function.
`These investigators also reported that such increased sensitivity to the drug could be produced
`in patients with normal renal function by administration of urea before thiopental. We were
`able to demonstrate the same dose-sparing effect of urea on thiopental in normal rats and we
`established that its mechanism was pharrnacokinetic rather than pharmacodynamic (Danhof
`and Levy, 1985). In further explorations on rats with either surgically or chemically-induced
`renal failure, we found that the concentrations of barbiturates required to produce sleep (loss
`of righting reflex) in rats were substantially decreased by renal failure (Danhof, Hisaoka and
`Levy, 1984). This pharmacodynamic alteration is mediated by an endogenous, dialyzable
`material which accumulates in renal failure (Hisaoka and Levy, 1985). Clearly, the ability to
`discover such effects of disease on drug action in preclinical studies is of enonnous value.
`Another example of the potential utility of preclinical pharrnacodynamic investigations
`derives from a tragic occurrence following the Japanese attack on Pearl Harbor during World
`
`10
`
`11
`
`

`

`War II. Surgery had to be performed on many casualties whose blood volume w
`as severely
`depleted by hemorrhage. The general anesthetic agent used in these procedures w
`as usua ly
`1
`thiopental. As reported by Halford in 1943, "death ensued in enough cases to cau
`, se us to
`abandon it (thiopental) as too dangerous". We found in studies on rats that heinor h
`.
`r ag1c
`.
`.
`.
`.
`hypovolemta caused a pronounced mcrease m central nervous system sensitivity to the
`depressant effect of a barbiturate, an observation which, had it been made before the use of
`thiopental at Pea~l H~rbor,_might have saved h~ndreds oflives (Klockowski and Levy, 1988).
`Theophyllme 1s a widely used bronchod1lator. Its usual therapeutic plasma concentra(cid:173)
`tion range is quite narrow and is frequently exceeded due to pronounced inter- and intraindi(cid:173)
`vidual variations in the total clearance of this drug. High concentrations of theophylline can
`cause neurotoxicity of which the most serious manifestation is generalized seizures that are
`often fatal or the cause of permanent brain damage. Noting that some patients had seizures at
`relatively low (but super therapeutic) plasma theophylline concentrations whereas others did
`not experience seizures even at ten-fold higher concentrations, we explored various potential
`risk factors for theophylline neurotoxicity in rats. We found one such risk factor to be renal
`failure; rats with bilaterally ligated ureters had convulsions at drug concentrations in cerebro(cid:173)
`spinal fluid that were about one-half those in normal controls (Ramzan and Levy, 1987).
`Significantly, Aitken and Martin (1985) concluded from a retrospective review of hospital
`records of patients with theophylline toxicity that "patients with renal dysfunction may be at
`higher risk for life-threatening complications of theophylline intoxication". This important
`observation could have been predicted (and lives could have been saved!) by appropriate
`preclinical studies.
`As a final example of the value of preclinical pharmacodynamics for predicting effects
`of underlying diseases in humans, I cite our experience with the benzodiazepines. Recent
`clinical reports have described pronounced increases in the central nervous system depressant
`effect of a benzodiazepine in hepatic cirrhosis compared to the effect on individuals without
`liver disease at comparable plasma concentrations of unbound drug (Fisch and co-workers
`'
`1986; Bakti and co-workers, 1987). We studied the pharmacokinetics and pharmacodynamics
`of desmethyldiazepam in rats with carbon tetrachloride induced liver dysfunction (Klock(cid:173)
`owski and Levy, to be published). Using rota rod performance as an index of CNS functionality,
`we observed that a serum concentration of about 0.04 mg/1 of unbound desmethyldiazepam
`produced an impairment of about 20 percent in normal rats and of 100 percent in rats with
`liver disease. Again, the preclinical observations could have predicted the clinical effects and
`prevented many cases of unanticipated adverse effects due to benzodiazepines in patients with
`liver disease.
`In conclusion, knowledge derived from preclinical pharmacodynamic studies on suit(cid:173)
`able animal models can be extraordinarily useful for the design and efficient perfonnance of
`subsequent clinical studies, for the development of phannacodynamic models and for antici(cid:173)
`pating and exploring the effects of variables such as underlying diseases on drug concentration
`vs. pharmacologic activity relationships. Drug development will be facilitated and human
`pharmacotherapy will become safer and more effective if properly conducted preclinical
`phannacodynamic studies become an integral part of the drug development process.
`
`11
`
`12
`
`

`

`REFERENCES
`
`Aitken, M. L., and T. R. Martin ( 1985). Life-threatening complications of theophylline
`toxicity are not predicted by serum theophylline levels. Amer. Rev. Resp. Dis., ill,
`A68.
`Bakti, G., H. U. Fisch, G. Karlaganis, C. Minder, and J. Bircher (1987). Mechanism of the
`excessive sedative response of cirrhotics to benzodiazepines: model experiments with
`triazolam. Hepatology, 1, 629-638.
`Cheung, W. K., L. L. Sia, D. L. Woodward, J. F. Graveline, R. E. Desjardins, A. Yacobi, and
`B. M. Silber (1988). Importance of oral dosing rate on the hemodynamic and pharma(cid:173)
`cokinetic profile on nilvadipine. J. Clin. Pharmaco)., 2.8., 1000-1007.
`Danhof, H., M. Hisaoka, and G. Levy (1984). Kinetics of drug action in disease states. II.
`Effect of experimental renal dysfunction on phenobarbital concentrations in rats at onset
`of loss of righting reflex. J. Pharmacol. Exp. Ther., 230, 627-631.
`Danh of, M., and G. Levy ( 1984). Kinetics of drug action in disease states. I. Effect of infusion
`rate on phenobarbital concentrations in serum, brain and CSP of normal rats at onset of
`loss of righting reflex. J. Pharmacol. Exp. Tuer., 229, 44-50.
`Danhof, M., and G. Levy (1985). Kinetics of drug action in disease states. V. Acute effect of
`urea infusion of phenobarbital concentrations in rats at onset of loss of righting reflex.
`J. Phannacol. Exp. Ther., 232, 430-444.
`Danhof, M., M. Hisaoka, and G. Levy (1985). Kinetics of drug action in disease states XII:
`Effect of experimental liver diseases on the pharmacodynamics of phenobarbital and
`ethanol in rats. J. Pharm. Sci,, 14., 321-324.
`Dundee, J. W., and R. K. Richards ( 1954). Effect of azotemia upon the action of intravenous
`barbiturate anesthesia. Anesthesiology, 15., 333-346.
`Fisch, H. U., G. Baktir, G. Karlaganis, and J. Bircher (1986). Excessive effects of ben(cid:173)
`zodiazepines in patients with cirrhosis of the liver: a phannacodynamic or a pharma(cid:173)
`cokinetic problem? Phannacopsychiatry, 19, 14.
`Francis, E. Z., C. A. Kimmel and D. C. Rees (1990). Workshop on the qualitative and
`quantitative comparability of human and animal developmental neurotoxicity: Sum(cid:173)
`mary and implications. NeurotoxicoI. Terato)., 12, 285- 292.
`Gianni, L., L. Vigano, A. Surbone, D. Ballinari, P. Casali, C. Tarella, and J.M. Collins (1990).
`Pharmacology and clinical toxicity of 4'-iodo-4'- deoxydoxorubicin: An example of
`successful application of pharmacokinetics to dose escalation in phase I trials. J. Natl.
`Cancer Inst., 82, 469-477.
`Halford, F. J. (1943). A critique of intravenous anesthesia in war surgery. Anesthesiologx, 4,
`67-69.
`Hendrie, T. A., J. R. Rowland, P. E. Binkerd, and A.G. Hendrickx (1990). Developmental
`toxicity and pharmacokinetics of phenytoin in the rhesus macaque: An interspecies
`comparison. Reproductive Toxicol., 1:, 257-266.
`Hisaoka, M., and G. Levy (1985). Kinetics of drug action in disease states. XIII. Effect of
`dialyzable component(s) of uremic blood on phenobarbital concentrations in rats at
`onset of loss of righting reflex. J. Pharmaco). Exp. Ther., 2.34, 180-183.
`Klockowski, P., and G. Levy (1988). Kinetics of drug action in disease states. XXIII. Effect
`of acute hypovolemia on the pharmacodynamics of phenobarbital in rats. J. Ph arm. Sci.,
`77, 365-366.
`
`12
`
`13
`
`

`

`Kutt, H., W. Winters, R. Kolenge, and F. McDowell (1964). Diphenylhydantoin metabolism,
`blood levels and toxicity. Arch. Neuro)., ll, 642-648.
`Levy, G. ( 1985). Variability in animal and human pharmacodynamic studies. In M. Rowland,
`L. B. Sheiner, and J.-L. Steimer (Eds.), Variability in Drug Therapy: Description,
`Estimation, and Control, Raven Press, New York, pp. 125-138.
`Nagashima, R., R. O'Reilly, and G. Levy (1969). Kinetics of pharmacologic effects in man:
`The anticoagulant action of warfarin. Clin. Pharmacol. Ther., 10, 22-35.
`Ramzan, I. (1990). Pharmacodynamics of phenytoin-induced ataxia in rats. Epilepsy Res.,
`.5., 80-83.
`Ramzan, I. M., and G. Levy (1987). Kinetics of drug action. XVIII. Effect of experimental
`renal failure on the pharmacodynamics of theophylline-induced seizures in rats. L
`Pharmacol. Exp. Ther., 240, 584-588.
`Routledge, P.A., P.H. Chapman, D. M. Davies, and M. D. Rawlins (1979). Pharmacokinetics
`and phannacodynamics of warfarin at steady state. Br. J. Clio. Pharmaco!., .S, 243-247.
`Wu, W-H., B. M. Henderson, R. Lane, D. Garnes, A. Yacobi and B. M. Silber (1988).
`Relationship between the pharmacokinetic and pharmacodynamic profile of nilvadip(cid:173)
`ine in the dog. Drug Metab. Dispos., 16, 222-227.
`Yacobi, A., and G. Levy (1977). Comparative phannacokinetics of coumarin anticoagulants
`XXVIII: predictive identification of rats with relatively steep serum warfarin concen(cid:173)
`tration-anticoagulant response characteristics. J. Pbmm. Sci., fi.6, 145.
`Yacobi, A., L. B. Wingard, Jr., and G. Levy (1974). Comparative pharmacokinetics of
`coumarin anticoagulants X: relationship between distribution, elimination, and antico(cid:173)
`agulant action of warfarin. J. Phann. Sci., fil, 868-872.
`
`This material wasc,ooied
`
`13
`
`14
`
`

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