throbber
Advancesin
`
`ENZYME REGULATION
`
`Volume 22
`
`Proceedings of the Twenty-Second Symposium on Regulation of Enzyme
`Activity and Synthesis in Normal and Neoplastic Tissues
`held at Indiana University School of Medicine
`Indianapolis, Indiana
`October 3 and 4, 1983
`
`Edited by
`GEORGE WEBER
`
`Indiana University School of Medicine
`Indianapolis, Indiana
`
`Technical editor
`Catherine E. Forrest Weber
`
`
`
`PERGAMONPRESS
`OXFORD - NEW YORK - TORONTO
`SYDNEY - PARIS : FRANKFURT
`
`1 of 32
`
`Alkermes, Ex. 1043
`
`1 of 32
`
`Alkermes, Ex. 1043
`
`

`

`UK.
`
`US.A.
`
`CANADA
`
`AUSTRALIA
`
`FRANCE
`
`Pergamon Press Ltd., Headington Hill Hall,
`Oxford OX3 OBW,England
`Pergamon Press Inc., Maxwell House, Fairview Park,
`Elmsford, New York 10523, U.S.A.
`PergamonPress Canada Ltd., Suite 104, 150 Consumers Road,
`Willowdale, Ontario M2J 1P9, Canada
`PergamonPress (Aust.) Pty. Lid., P.O. Box 544,
`Potts Point, N.S.W. 2011, Australia
`Pergamon Press SARL, 24 rue des Ecoles,
`75240 Paris, Cedex 05, France
`
`FEDERAL
`REPUBLIC OF
`GERMANY
`elma
`Copyright © 1984 Pergamon Press Ltd.
`
`Pergamon Press GmbH, Hammerweg 6, D-6242
`Kronberg-Taunus, Federal Republic of Germany
`
`All Rights Reserved. No part of this publication may be
`in
`reproduced, stored in a retrieval system, or transmitted,
`—
`any form or by any means, electronic, electrostatic, magnenc
`tape, electronic, mechanical, photocopying, recording or otherwise,
`without permission in writing from the publishers.
`First edition 1984
`
`Library of Congress Catalog Card No. 63-19609
`
`ISBN 0 08 031498 &
`ISSN 0065-2571
`
`Printed in Great Britain by A. Wheaton & Co. Lid., Exeter
`
`2 of 32
`
`Alkermes, Ex. 1043
`
`2 of 32
`
`Alkermes, Ex. 1043
`
`

`

`Advances in
`
`ENZYME REGULATION
`
`Volume 22
`
`3 of 32
`
`Alkermes, Ex. 1043
`
`3 of 32
`
`Alkermes, Ex. 1043
`
`

`

`QUANTITATIVE ANALYSIS OF
`DOSE-EFFECT RELATIONSHIPS: THE
`COMBINED EFFECTS OF MULTIPLE
`DRUGS OR ENZYME INHIBITORS
`
`TING-CHAO CHOU* and PAUL TALALAYT
`
`*Laboratory of Pharmacology, Memorial Sloan-Kettering Cancer Center, New York, NY
`10021, and {Department of Pharmacology and Experimental Therapeutics, The Johns
`Hopkins University School of Medicine, Baltimore, Maryland 21205
`
`INTRODUCTION
`The quantitative relationship between the dose or concentration of a given
`ligand andits effect is a characteristic and important descriptor of many
`biological systems varying in complexity from isolated enzymes(or binding
`proteins)
`to intact animals. This
`relationship has been analyzed in
`considerable detail for reversible inhibitors of enzymes. Such analyses have
`made
`assumptions
`on
`the mechanism of
`inhibition (competitive,
`noncompetitive, uncompetitive), and on the mechanism ofthe reaction for
`multi-substrate enzymes (sequential or ping-pong), and have required
`knowledge of kinetic constants (1-4). More recently, it has been possible to
`describe the behavior of such enzyme inhibitors by simple generalized
`equations that are independent of inhibitor or reaction mechanisms and do
`not require knowledge of conventional kinetic constants (i.e. K,,, Kj, Vinay)
`(5-8).
`Our understanding of dose-effect relationships in pharmacological systems
`has not advanced to the samelevel as those of enzyme systems. Manytypes of
`mathematical transformations have been proposedto linearize dose-effect
`plots, based on statistical or empirical assumptions,e.g. probit (9, 10), logit
`(11) or power-law functions (12). Although these methods often provide
`adequatelinearizations ofplots, the slopes and intercepts of such graphsare
`usually devoid of any fundamental meaning.
`
`THE MEDIAN EFFECT PRINCIPLE
`We demonstrate here the application ofa single and generalized method for
`analyzing dose-effect relationships in enzymatic, cellular and whole animal
`systems. Wealso examine the problem of quantitating the effects of multiple
`inhibitors on such systems and provide definitions of summationofeffects,
`and consequently of synergism and antagonism.
`Since the proposed method ofanalysis is derived from generalized mass
`action considerations, we caution the readerthat the analysis of dose-effect
`2
`AER 22-8"
`27
`
`4 of 32
`
`Alkermes, Ex. 1043
`
`4 of 32
`
`Alkermes, Ex. 1043
`
`

`

`28
`
`TING-CHAO CHOU and PAUL TALALAY
`
`data is concerned with basic mass-action characteristics rather than with
`proof of specific mechanisms. Nevertheless, it is convenient and intuitively
`attractive to analyze and normalize all types of dose-response results by a
`uniform method which is based on sound fundamental considerations that
`have physicochemical and biochemical validity in simpler systems. Our
`analysis is based on the median effect principle of the mass action law (5-8),
`and has already been showntobesimple to apply and useful in the analysis of
`complex biological systems (13).
`
`The Median Effect Equation
`The median effect equation (6, 8) states that:
`
`f/f, = (D/D,)"
`
`(1)
`
`where D is the dose, f, and f, are the fractions of the system affected and
`unaffected, respectively, by the dose D, D,, is the dose required to produce the
`median effect (analogous to the morefamiliar IC), EDs), or LD, values), and
`m is a Hill-type coefficient signifying the sigmoidicity ofthe dose-effect curve,
`i.c.,m = 1 for hyperbolic (first order or Michaelis-Menten)systems. Since by
`definition, f, + f, = 1, several useful alternative forms of equation | are:
`
`f,/0 he f,) = ((f,y" a iT re [(f,y" = 1)] = (D/D,,)"
`
`f, = 1/(1 + (D,/D)"
`
`p= D,[f,/01 ~ jy"
`
`The median effect equation describes the behavior of many biological
`systems.It is, in fact, a generalized form of the enzyme kinetic relations of
`Michaelis-Menten (14) and Hill (15), the physical adsorptionisotherm of
`Langmuir (16), the pH-ionization equation of Henderson and Hasselbalch
`(17),
`the equilibrium binding equation of Scatchard (18), and the
`pharmacological drug-—receptor interaction (19). Furthermore, the median
`effect equation is directly applicable not only to primary ligands such as
`substrates, agonists, and activators, but also to secondary ligands such as
`inhibitors, antagonists, or environmental factors (5, 6).
`When applied to the analysis of the inhibition of enzyme systems, the
`median effect equation can be used without knowledge of conventional
`kinetic constants(i.e. K,,, V,,,, or K,) and irrespective of the mechanismof
`inhibition(i.e. competitive, noncompetitive or uncompetitive). Furthermore,
`itis valid for multisubstrate reactions irrespective of mechanism (sequential or
`ping-pong) (5-8).
`
`5 of 32
`
`Alkermes, Ex. 1043
`
`5 of 32
`
`Alkermes, Ex. 1043
`
`

`

`ANALYSIS OF MULTIPLE DRUG EFFECTS
`
`29
`
`The Median Effeet Plot
`The median effect equation (equation |) may be linearized by taking the
`logarithms ofboth sides, i.e.
`
`or
`or
`
`log (f,/f,) = m log (D) - m log (D,,)
`log [(f,)"' - 1]"' = m log (D) - mlog (D,,)
`log f(f," ~ 1] =m log (D) -m log (D,,)
`
`The median effect plot (Fig. 1) of y = log (£,/f,) or its equivalents with
`respect to x = log (D) is a general and simple method (13, 30) for determining
`pharmacological median doses for lethality (LD,9), toxicity (TD,,), effect of
`agonist drugs (ED,,), andeffect of antagonist drugs (IC,,). Thus, the median-
`effect principle of the mass-action law encompasses a wide range of
`applications. The plot gives the slope, m, and the intercept ofthe dose-effect
`plot with the median-effect axis [i.e. when f, = f,, f/f, = 1 and hence y = log
`(f,/f,) = 0] which gives log (D,,) and consequently the D,, value. Any cause-
`consequencerelationship that gives a straightline for this plot will provide the
`two basic parameters, m and D,,, and thus, an apparent equation that
`describes such a system. Thelinearity of the median-effect plot (as determined
`from linear regression coefficients) determines the applicability of the present
`method.
`
`
`
`
`
`Log[(fa)-!-1]"
`
`
`
`Log (D/Dm)
`
`FIG. 1. The median-effect plot at different slopes correspondingto m valuesof0.5, 1, 2and 3. The
`plot
`is based on the median-effect equation (equation 1)
`in which the dose (D) has been
`normalized by taking the ratio to the median-effect dose (D,). Note that the ordinatelog[(f, yi-
`iy! is identical to log [f,y' — 1] or log (f,/fy).
`
`6 of 32
`
`Alkermes, Ex. 1043
`
`6 of 32
`
`Alkermes, Ex. 1043
`
`

`

`30
`
`TING-CHAO CHOU and PAUL TALALAY
`
`Relation of the Median-Effect Equation to Michaelis-Menten and Hill
`Equations
`In the special case, when m = 1, equation | becomes f, = [1 + (D,,/DT'
`which has the sameform as the Michaelis-Menten equation (14), v/V,,,. =[1 +
`(K,,/S)}T'. In addition, when the effector ligand is an environmental factor
`suchas an inhibitor, the equation, f, = [1 +(D,,/D)f', is valid not only fora
`single substrate reaction (Michaelis-Menten equation) but also for multiple
`substrate reactions;
`the fractional effect
`is expressed with respect
`to the
`control velocity rather than to the maximal velocity (6). Furthermore, f, in
`equation |
`is simple to obtain, whereas the determination ofV,,,, in the
`Michaelis-Menten (or Hill) equations requires approximation or extra-
`polation (6, 7). The logarithmic form of equation | describes the Hill equation.
`
`The Utility of the Median Effect Principle
`The median-effect equation has been used to obtain accurate values of ICsp,
`ED.,, LDso, or the relative potencies of drugs or inhibitors in enzyme systems
`(6-8, 21-26), in cellular systems (20, 27, 28) and in animal systems (13, 29-32).
`Analternative form of the median-effect equation (5) has been used for
`calculating the dissociation constant (K, or K,) of ligands for pharmacological
`receptors
`(33-35),
`It has
`also permitted the
`analysis of chemical
`carcinogenesis data and haspredicted especially accurately tumorincidenceat
`low dose carcinogen exposure (30, 31). By using the median-effect principle,
`the general equation for describing a standard radioimmunoassayorligand
`displacementcurve has been derived recently by Smith (36). It has also been
`used to showthat there is marked synergism among chemotherapeutic agents
`in the treatment of hormone-responsive experimental mammary carcinomas
`(32).
`In recent preliminary reports (13, 37), we have shown that,
`in
`conjunction with the multiple drug effect equations (see below), the median-
`effect plot formsthe basis for the quantitation of synergism, summationand
`antagonism of drugeffects.
`
`ANALYSIS OF MULTIPLE DRUG EFFECTS
`
`An Overview
`
`the past decades, numerous authors have claimed synergism,
`Over
`summation or antagonism oftheeffects of multiple drugs. However,thereis
`still no consensusas to the meanings ofthese terms. For instance, in a review,
`Goldin and Mantel
`in 1957 (38) listed seven different definitions of these
`terms. Confusion and ambiguity persist (39) despite increasing use ofmultiple
`drugs in experimentation and in therapy. This emphasizes the lack ofa
`
`7 of 32
`
`Alkermes, Ex. 1043
`
`7 of 32
`
`Alkermes, Ex. 1043
`
`

`

`ANALYSIS OF MULTIPLE DRUG EFFECTS
`
`31
`
`theoretical basis that would permit rigorous and quantitative assessment of
`the effects of drug combinations.
`Attemptsto interpret the effect of multiple drugs have been documented for
`more than a century (39). Since the introduction of the isobol concept by
`Loewe in 1928 (40, 41) and the fractional product concept (see Appendix) by
`Webbin 1963 (42), the theoretical and practical aspects of the problem have
`been the subject of many reviews (38, 43-51). Some authors have discussed the
`possible mechanisms that may lead to synergism, and others have emphasized
`methods of data analysis. The kinetic approach was used earlier by some
`investigators (4, 42, 52-58), but the formulations were frequently too complex
`to be of practical usefulness or were restricted to individual situations.
`Although not specifically stated, some formulations are limited to two
`inhibitors; others are valid only for first order (Michaelis-Menten type)
`systems but not for higher order (Hill type) systems, andstill others are valid
`only for mutually nonexclusive inhibitors but not for mutually exclusive
`inhibitors.
`The present authors, therefore, have undertaken a kinetic approach to
`analyze the problem. An unambiguous definition of summation is a
`prerequisite for any meaningful conclusions with respect to synergism and
`antagonism. Ironically, two prevalent concepts for calculating summation
`i.e., the isobol and the fractional-product method, are shown to conform to
`two opposite situations. The former conceptis valid for drugs whose effects
`are mutually exclusive, and thelatter is valid for mutually nonexclusive drugs
`(13, 49), and thus these methods cannot be used indiscriminately (see
`Appendix). In this paper, we provide the equations for both situations and
`show that they are merely special cases of the general equations described
`recently (59). We also propose a general diagnostic plot to determine the
`applicability of experimental data, to distinguish mutually exclusive from
`nonexclusive drugs, and to obtain parametersthat can bedirectly used for the
`analysis of summation, synergism or antagonism.
`
`Requirements for Analyzing Multiple Drug Effects
`The following informationis essential for analyzing multiple drug effects
`and for quantitating synergism, summation and antagonism of multiple
`drugs.
`1. A quantitative definition of summation is required since synergism
`implies more than summation and antagonismless than summationofeffects.
`2, Dose-effect relationships for drug 1, drug 2 and their mixture (at a known
`ratio of drug 1 to drug 2) are required.
`a. Measurements madewith single doses of drug |, drug 2 and their mixture
`can never alone determine synergism since the sigmoidicity of dose-effect
`
`8 of 32
`
`Alkermes, Ex. 1043
`
`8 of 32
`
`Alkermes, Ex. 1043
`
`

`

`32
`
`TING-CHAO CHOU and PAUL TALALAY
`
`curves and the exclusivity of drug effects cannot be determined from such
`measurements.
`relationships should follow the basic mass-action
`b. The dose-effect
`principle relatively well (e.g. median-effect plots with correlation coefficients
`for the regression lines greater than 0.9).
`c. Determination of the sigmoidicity of dose-effect curves and the
`exclusivity ofeffects of multiple drugs is necessary. The slope of the median-
`effect plot gives a quantitative estimation of sigmoidicity. When m= 1, the
`dose-effect curve is hyperbolic, when m # 1,
`the dose-effect curve is
`sigmoidal, and the greater the m value, the greater its sigmoidicity; m< lisa
`relatively rare case whichin allosteric systems indicates negative cooperativity
`of drug binding at the receptor sites. When the dose-effect relationships of
`drug 1, drug 2 and their mixture areall parallel in the median-effect plot, the
`effects of drug | and drug 2 are mutually exclusive (59). If the plots of drugs |
`and 2 are parallel but the plot of their mixture is concave upward with a
`tendencytointersect the plot of the more potent ofthe twodrugs, their effects
`are mutually nonexclusive (59). If the plots for drugs | and 2 and their mixture
`are not parallel to each other, exclusivity of effects cannot be established.
`Alternatively, exclusivity of effects may not be ascertained because ofa
`limited numberof data points or limited dose range. In these cases, the data
`may be analyzed for the ‘combination index”(see below) on the basis of both
`mutually exclusive and mutually nonexclusive assumptions. Note that
`exclusivity may occur at a receptorsite, at a point in a metabolic pathway, or
`in more complex systems, depending on the endpoint of the measurements.
`
`Equations for the Effects of Multiple Drugs
`A systematic analysis in enzymekinetic systemsusing the basic principles of
`the mass action law has led to the derivation of generalized equations for
`multiple inhibitors or drugs (8, 59).
`
`1. For two mutually exclusive drugs that obey first order conditions. If two
`drugs(e.g., inhibitors D, and D,) haveeffects that are mutually exclusive, then
`the summation of combinedeffects(f,), 9, in first-order systems(i.c., each drug
`follows a hyperbolic dose-effect curve) can be calculated from (59):
`
`(f.)i.2 say
`(fwi.2
`
`(fi
`(fur
`
`
`4 (fi
`(fue
`
`
`(D),
`(EDs),
`
`mn
`
`(D),
`(EDs)
`
`(2)
`
`9 of 32
`
`Alkermes, Ex. 1043
`
`9 of 32
`
`Alkermes, Ex. 1043
`
`

`

`ANALYSIS OF MULTIPLE DRUG EFFECTS
`
`33
`
`wheref, is the fraction affected and f, is the fraction unaffected, and EDsois
`the concentration of the drug that is required to produce a 50% effect. Note
`that f, +f, = l orf, =1-f,.
`
`2. For two mutually nonexclusive drugs that obeyfirst order conditions. If the
`effects of two drugs (D, and D,) are mutually non-exclusive(i.e., they have
`different modes ofaction or act independently) the summation of combined
`effects, (f,),2, in a first-order system is (59):
`
`(io _ Gy, Ge, ah
`(fy).
`(f.)
`(f)
`(fs (Gu)
`
`
`_ (D),
`(Dy, Di )
`
`
`
`(EDy) (ED~(ED) (ED 3)
`
`involving more than two
`relationships apply to situations
`Similar
`inhibitors, for which generalized equations are given in ref. 59. In enzyme
`systems, equations 2 and 3 express summation of inhibitory effects,
`irrespective of the number of substrates,
`the type or mode ofreversible
`inhibition (competitive, noncompetitive or uncompetitive) or the kinetic
`mechanisms (sequential or ping-pong) of the reaction under consideration.
`The simplicity of the above equations(in whichall specific kinetic constants,
`substrate concentration factors, and V,,,, have been cancelled out during
`derivation) suggests their general applicability (5, 6). This is in contrast to the
`mechanism-specific reactions (3, 5) for which the equations are far more
`complex.
`In more organized cellular or animal systems,
`the dose-effect
`relationships of drugs or inhibitors are frequently sigmoidal rather than
`hyperbolic.
`
`3. For two mutually exclusive drugs that obey higher order conditions. The
`above concepts have been extended to higher-order (Hill-type) systems in
`which each drug has a sigmoidal dose-effect curve (i.e., has more than one
`binding site or exhibits positive or negative cooperativity). If the effects of
`such drugs are mutually exclusive:
`
`(fi
`i =
`(fia
`(of Lh
`
`‘i
`
`+
`
`a
`
`(fide
`(fi)
`
`(D),
`a
`i (D),
`
`(EDs)
`(EDso
`
`(gy
`
`where m is a Hill-type coefficient which denotes the sigmoidicity of the
`dose-effect curve.
`
`10 of 32
`
`Alkermes, Ex. 1043
`
`10 of 32
`
`Alkermes, Ex. 1043
`
`

`

`34
`
`TING-CHAO CHOU and PAUL TALALAY
`
`4. For two mutually nonexclusive drugs that obey higher order conditions. \f
`the effects of two drugs (D, and D,) are mutually nonexclusive andif each
`drug and their combination follow a sigmoidal dose-effect relationship with
`m" order kinetics, then this relationship becomes (59):
`
`®ialz. Texls,ee] =,fewery=
`
`
`
`a Beane +|a Fe en
`(fia
`(f,);
`(f.,)p
`(fi), (f,)
`
`
`(D);
`‘.
`(D);
`(EDs),
`(EDs),
`
`(D), (D),
`(EDs); (EDso),
`
`(5)
`
`In the special case where (f,),» = (f,),2 = 0.5, equations 2 and 4 become:
`
`(Dy, Db
`(ED.),
`(EDs)
`
`-
`
`(6)
`
`which describes the ED.) isobologram.
`Similarly, equations 3 and 5 become:
`
` (Dy, @h
`-
`(D) (D):
`
`(ED),~~(ED) (EDs), (ED); (7
`
`
`
`which doesnot describe an isobologram,because ofthe additional term on the
`left.
`is shown that equation 3 or 7 can be readily used for
`In the Appendix it
`deriving the fractional product equation of Webb (42), and equation4 can be
`used for deriving the generalized isobologram equation for any desiredf,
`value. Thus, for the isobologram at anyfractional effect f, = x per cent, the
`generalized equation is:
`
`
`Dy , Ox _
`(D,);
`(D,)
`
`(8)
`
`The limitations of the fractional product concept and the isobologram
`methodare detailed in the Appendix.
`
`11 of 32
`
`Alkermes, Ex. 1043
`
`11 of 32
`
`Alkermes, Ex. 1043
`
`

`

`35
`ANALYSIS OF MULTIPLE DRUG EFFECTS
`5. Quantitation of
`synergism,
`summation and antagonism. When
`experimentalresultsare entered into equations 2-5, ifthesu bef two terms
`in equation 2 or4, or the sum ofthe three terms a equation 3 or 5 is greater
`than, equal
`to, or smaller than 1,
`jt may be inferred that qntsaoniin
`summation or synergism ofeffects, respectively, has been observed. There-
`fore, from equations 2-5, if the combined obse
`‘ect
`is
`r
`calculated additive effect, (f.),,
`tved effect is greater than the
`2» Synergism is indicated;
`if
`it
`is
`smé
`eres
`d;
`is smaller,
`if it
`antagonism is indicated.
`It
`is, however, convenient to designate a
`combination index” (CI) for
`Bere
`:
`‘
`quantifying synergism, summation » and antagonism, as follows:
`
`cr= @h , Oy
`(D,);
`(D,);
`
`0)
`
`for mutually exclusive drugs, and
`
`= © , © , OM
`
`
`(D.),=(Dy)~~(D,), (D),); (10)
`
`for mutually nonexclusive drugs,
`For mutually exclusive or nonexclusive drugs,
`when CI<I, synergism is indicated.
`CI = 1, summation is indicated.
`CI > 1, antagonism is indicated,
`To determine synergism, summation and antagonism at any effect level
`(i.e., for any {, value), the procedure involves three steps: i) Construct the
`median-effect plot (Eqn. 1) which determines m and D,, values for drug 1,
`drug 2 and their combination; ii) for a given degreeofeffect(i-e., a givenf,
`value representing x per cent affected), calculate the corresponding doses[i.e.,
`(D,),, (D,) and (D,),2] by using the alternative form of equation 1, D, = D,,
`[£,/(1 - £,)}'"; tii) calculate the combinationindex (CI) by using equations 9 or
`10, where (D,), and (D,), are fromstep(ii), and (D,), 5 [also fromstep(ii)] can
`be dissected into (D), and (D), by their knownratio, P/Q. Thus, (D), =(D,)}»
`« P/(P + Q) and (D), = (D2 X Q/(P + Q), CI values that are smaller than,
`equalto, or greaterthan 1, represent synergism, summation and antagonism,
`respectively.
`To facilitate the calculation, a computer program written in BASIC for
`automatic graphing of CI with respect to f, has been developed. Samples of
`this computer simulation are shown in the examples to be given later. A
`sample calculation of CI without using a computeris also given in Example 1.
`
`12 of 32
`
`Alkermes, Ex. 1043
`
`12 of 32
`
`Alkermes, Ex. 1043
`
`

`

`36
`
`TING-CHAO CHOU and PAUL TALALAY
`
`APPLICATIONS OF THE MEDIAN EFFECT EQUATION
`AND PLOTS TO THE ANALYSIS OF MULTIPLE
`DRUGS OR INHIBITORS
`
`Example 1. Inhibition of Alcohol Dehydrogenase by Two Mutually Exclusive
`Inhibitors
`Yonetani and Theorell (55) have reported the inhibition of horse liver
`alcohol dehydrogenase by twoinhibitors (I, = ADP-ribose and I], = ADP)
`both of which are competitive with respect to NAD. Velocity measurementsin
`the presence of a range of concentrations of the two inhibitors (alone and in
`combination) and control velocities were retrieved from the original plot, and
`tabulated in ref. 59. The results are most conveniently expressed as fractional
`velocities (f,) which are the ratios of the inhibited velocities to the control
`velocities, and therefore correspondto the fraction of the process unaffected
`(f,). The fractional velocities in the presence of ADP-ribose (95-375 um), ADP
`(0.5-2.5 um), and a combination of ADP-ribose and ADP ata constant molar
`ratio of 190:1, havebeen plotted as log [(f,)' - 1] with respectto log (I) (Fig.2).
`For ADP-ribose, m = 0.968, I) = 156.1 uM with a regression coefficient of r=
`0.9988. For ADP, m = 1.043, Ij) = 1.657 wm and r = 0.9996, For ADP-ribose
`and ADPin combination (molarratio 190:1), m,= 1.004, (Ix);» = 107.0 um
`and r=0,9997.It is clear that both inhibitorsfollow first-order kinetics(i.e., m
`= 1) and that ADP-ribose and ADP are mutually exclusive inhibitors (i.e., the
`
`1,: ADPR

`T,: AOP
`o
`@ 1,41): 190:
`
`2
`
`i
`
`Itl,
`
`I,
`
`20
`
`i)
`
`= 10
`
`o5
`
`°o
`
`-05
`
`a=
`
`a
`2
`
`
`J
`l
`ar.
`_L
`1
`i
`!
`-0.5
`0
`0.5
`lo
`0.5
`2.0
`2.5
`30
`log (1)
`
`FIG. 2, Median-effect plots of the experimental data of Yonetani and Theorell (55) for the
`inhibition of horseliver alcohol dehydrogenase by two mutually exclusive inhibitors. 1,
`is ADP-
`ribose (ADPR), I, is ADP, and 1, + I, is a mixture of ADP-ribose and ADPin a molar ratioof
`190:1. The abscissa represents log (1); (@), log (Lp (0), or log [(1)) + (Dp (19051; A). In this case itis
`convenient to use the terms fractional velocity (f,) whichis the ratio ofthe inhibited to the control
`velocity and therefore corresponds tothefraction that is unaffected ({,,). [from Chou andTalalay
`(59)].
`
`13 of 32
`
`Alkermes, Ex. 1043
`
`13 of 32
`
`Alkermes, Ex. 1043
`
`

`

`ANALYSIS OF MULTIPLE DRUG EFFECTS
`37
`plot for the combination of inhibitors parallels the plots for each of the
`component
`inhibitors). These conclusions are in agreement with the
`interpretations obtained by Yonetaniand Theorell (55) and Chou and Talalay
`(59) using different methods. For the present analysis, knowledge ofkinetic
`constants and type of inhibition is not required. The plots show excellent
`agreement between theory and experiment.
`‘with this knowledge of the m and Is) values for each inhibitor and the
`combination at a constant molar ratio, it is possible to calculate the inhibitor
`combination index (Cl) for a series of values of f, (Fig. 3). The Cl values are
`close to | over the entire range of f, values, suggesting strongly that the
`inhibitory effects of ADP-ribose and ADPare additive.
`3
`
`2
`
`1
`
`etrecemeerriees,
`
`spergmengteeepeeeeneweset*apecengeeneyteerysempeeenpeees
`
`Antagonistic
`
`4* Additive
`
`
`
`Synergistic
`
`FIG. 3. Computer-generated graphical presentation of the combination index (C1) with respect to
`fraction affected(f,) for the additive inhibition by ADP-ribose and ADP (molarratioof 190:1) of
`horse liver alcohol dehydrogenase. The plot is based on equation 9 (mutually exclusive) as
`described in the section entitled “Quantitation of Synergism, SummationandAntagonism.” Cl is
`the combinationindex whichis equal to (D)(D,); +(D) (Dp (see text for sample calculation).
`Cl < 1, = 1 and > |
`represent synergistic, additive and antagonistic effects, respectively.
`Althoughplots of Cliwith respect to f, can be obtained by step-by-step calculations,it is much
`more convenient to use computer simulation. The parameters were obtained as described in Fig.
`2, by the use of linear regression analysis or computer simulation.
`
`Wenow give a samplecalculation of the combination index (CI) for an
`arbitrarily selected value off, = 0.9:
`From equation 1, D, = D,, [f,/(1 - £,)]”.
`Since 1,
`is ADP-ribose and I,
`is ADP,
`then (Dy); = 156.1 um [0.9/1 - 0.9)]°* = 1511 um
`(Dy), = 1.657 xm [0.9/(1 - 0.9)77°" = 13.62
`(Dy)2 = 107.0 um [0.9/1 - 0.9)]'" = 954.6 uM
`
`14 of 32
`
`Alkermes, Ex. 1043
`
`14 of 32
`
`Alkermes, Ex. 1043
`
`

`

`38
`
`TING-CHAO CHOU and PAUL TALALAY
`
`therefore, (CD y° =
`
`Since in the mixture I, :1, = 190:1,
`then, (Dp), can be dissected into:
`(D), = 954.6 X [190/(190 + 1)] = 949.6 um
`(D), = 954.6 * [1/(190 + 1)] = 4.998 um
`949.6 uM ¥ 4.998 uM
`1511 um
`13.62 jum
`is close to 1, an additive effect of ADP-ribose and
`Since value of (CI)s,
`ADP at f, = 0.9 is indicated.
`A computer program for automated calculation of m, D,,, D,, r, and CI at
`different f, values has been developed.
`
`= 0.9955.
`
`Example 2. Inhibition of Alcohol Dehydrogenase by Two Mutually Non-
`Exclusive Inhibitors
`Yonetani and Theorell (55) also studied the inhibitionofhorse liver alcohol
`dehydrogenase by the two competitive, mutually nonexclusive inhibitors: o-
`phenanthroline (I,) and ADP (I,). The fractional velocity (f,) values retrieved
`from the original plot are given in ref. 59, and are presented in the form ofa
`median-effect plot, ie., log [(f,y' — 1] with respect
`to log (1) (Fig. 4). 0-
`Phenanthroline gives m = 1.303, Is) = 36.81 uM and r=0.9982, and ADP gives
`m = 1,187, Is) = 1.656 wm and r = 0.9842. These data again show that both
`inhibitors follow first order kinetics (i.e., m ~ 1). However, when the data for
`
`20
`
`: O- phenanthroline
`* I,
`* I, : ADP
`4 I,+1,: 17.4:1
`
`log[(fy)"'=1]
`
`-0.5
`
`oO
`
`0.5
`
`Lo
`
`1.5
`
`2.0
`
`2.5
`
`log (1)
`
`FIG. 4. The median-effect plot of experimental data of Yonetani and Theorell (55) for the
`inhibition ofhorse liver alcohol dehydrogenase by two mutually nonexclusive inhibitors, |; is o-
`phenanthroline, h is ADP, and }, + 1; isa mixture of o-phenanthroline and ADP (molar ratio
`17.4:1). The abscissa represents log (1), (@), log (Ip (©), or log [(1), + (Dp J (A) [from Chou and
`Talalay (59)].
`
`15 of 32
`
`Alkermes, Ex. 1043
`
`15 of 32
`
`Alkermes, Ex. 1043
`
`

`

`ANALYSIS OF MULTIPLE DRUG EFFECTS
`
`39
`
`the mixture of o-phenanthroline and ADP (constant molarratio 17.4:1) are
`plotted in the same manner, a very different result is obtained: m,, = 1.742
`(apparent), (sy), = 9.116 wm and r = 0.9999. The dramatic increase in the
`slope ofthe plot for the mixture (in comparison to each of its components),
`clearly indicates that o-phenanthroline and ADP are mutually nonexclusive
`inhibitors.
`levels are given in Figure 5. The
`The combination indices at various f,
`results indicate that there is a moderate antagonism at low f, values and a
`marked synergism at high f, values,
`
`3
`
`2
`
`cI
`
`fh
`
`‘
`*.
`Antagonisa
`s Sensis Roe teh nmenmel
`retry.
`
`Synergism
`
`FIG. 5, Computer-generated graphical presentation ofthe combinationindex (CI) with respect to
`fraction affected (f,) for the inhibition ofhorse liver alcohol dehydrogenase by a mixture ofo-
`phenanthroline and ADP (molar ratio 17.4:1). The method of analysis is the same as that
`described in the legend to Fig, 3, except that equation 10 (mutually non-exclusive) is used.
`
`Example 3. Inhibition ofthe Incorporation of Deoxyuridine into the DNA of
`L1210 Leukemia Cells by Methotrexate (MTX) and 1-B-p-
`Arabinofuranosyleytosine (ara-C)
`Murine L1210 leukemiacells were incubated in the presence of a range of
`concentrations of MTX (0.1-6.4 4), of ara-C (0.0782-5.0 uM), or a constant
`molar ratio mixture of MTX and ara-C (1:0.782), and the incorporation of
`deoxyuridine into DNA was then determined. Thefractional inhibitions (f,)
`of dUrd incorporation are shown in Table 1. Analysis of the results by the
`median effect plot (Fig. 6) gave the following parameters: for MTX, m =
`1.091, Dj, = 2.554 um, r= 0.9842; for ara-C, m= 1.0850, D,, = 0.06245 um, and
`
`16 of 32
`
`Alkermes, Ex. 1043
`
`16 of 32
`
`Alkermes, Ex. 1043
`
`

`

`40
`
`TING-CHAO CHOU and PAUL TALALAY
`
`TABLE 1. INHIBITION OF[6H]DEOXYURIDINE(dUrd) INCORPORATION INTO
`DNAIN L1210 LEUKEMIA CELLS BY METHOTREXATE (MTX) AND 1-B-p-
`ARABINOFURANOSYLCYTOSINE (ARA-C), ALONE AND IN COMBINATION
`
`Fractional inhibition (f,) at [ara-C| of
`0
`0.782
`0.156
`0.313
`0.625
`£25
`25
`5.0
`MTX
`
` MM uM BM uM eM BM EM
`
`
`
`
`
`BM
`
`0.582
`0.405
`
`O.715
`
`0.587
`
`0.860
`
`0.926
`
`0.955
`
`0.980
`
`0,993
`
`0
`0
`0.0348
`0.1
`ND*
`0.2
`ND
`0.4
`0.140
`0.8
`0.415
`1.6
`0.970
`0.573
`3.2
`
`
`0.7556.4 ND
`
`0.775
`
`O.878
`
`0,943
`
`*Result not used because of large variation between duplicates.
`L1210 murine leukemia cells (8 * 10° cells) were incubated in Eagle’s basal medium (20) in the
`presence and absence of various concentrations of MTX and ara-Candtheir mixture (molar
`ratio, 1:0.782) at 37°C for 20 min and then incubated with 0.5 wm (1 pCi) of [6-H ]JdUrd, at 37°C
`for 30 min. Fractional inhibition (f, or f,) of [6H ]dUrd incorporation into perchloric acid-
`insoluble DNA fraction was then measuredas previously described (20). All measurements were
`made in duplicate.
`
` Ara-C + MTX
`
`(0.782:1)
`
`eae mm
`
`wes miter
`
`nie ert
`
`nce
`
`-1
`
`o
`
`|
`
`2
`
`log
`
`(Concentration, 7M]
`
`_|
`3
`
`4
`
`9
`
`-l
`
`-2
`-2
`
`as
`y
`
`2=
`
`o2
`
`FIG. 6. Median-effect plot showing the inhibition of [(6~H]dUrd incorporation into DNA of
`L1210 murine leukemia cells by methotrexate (MTX), (©); arabinofuranosylcytosine (ara-C), (*);
`or their mixture (1:0.782), (+). Data from Table | have been used.
`
`r= 0.9995. For the combination of MTX and ara-C (1:0.782), the parameters
`were: m = 1.1296, D,, = 0.2496 wo, and r = 0.9995. The combination index
`(Fig. 7) shows a moderate antagonism between the two drugsatall values of
`fractional inhibition.
`
`17 of 32
`
`Alkermes, Ex. 1043
`
`17 of 32
`
`Alkermes, Ex. 1043
`
`

`

`ANALYSIS OF MULTIPLE DRUG EFFECTS
`
`41
`
`a
`
`be
`
`sg——
`
`cI
`
`-
`
`Neeeeeteesccecevetteteseg,
`
`Antagonism
`ol
`Synergism
`
`
`
`FIG, 7. Computer-generated graphical presentation ofthe drug combination index (CI) with
`respect to fraction affected(f{,) for the inhibitoryeffect of a mixture of methotrexate (MTX) and
`arabinofuranosyleytosine (ara-C) (molar ratio, 1:0.782) on the incorporation of[6-H]dUrd into
`DNA of L1210 murine leukemia cells. The data from Table | andthe parameters obtained from
`Fig. 6 have been used for this plot on the assumption that the drugs act in a mutually exclusive
`manner (equation 9).
`
`Example 4. Inhibition of the Incorporation of Deoxyuridine into the DNA of
`1.1210 Leukemia Cells by Hydroxyurea (HU) and 5-Fluorouracil (5-FU)
`Murine L1210 leukemia cells were incubated in the presenceof a range of
`concentrations of hydroxyurea (50-3,200 uM), or 5-fluorouracil (4.0-256 ym),
`and of a constant molar ratio mixture of hydroxyurea and 5-fluorouracil
`(12.5:1), and the incorporation of deoxyuridine (dUrd) into DNA was then
`determined. Thefractional inhibitions (f,) of dUrd incorporation are shown
`in Table 2. Analysis of the results by the median effect plot (Fig. 8) gave the
`following parameters: for hydroxyurea, m = 1.196, D,, = 34.09 um, and r=
`0.9908; for 5-fluorouracil, m= 1.187, D,, = 8.039 uM, and r= 0.9978; and for
`the mixture of hydroxyureaand5-fluorouracil (12.5:1), m= 1.407, D,, = 225.8
`po

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