throbber

`
`AAC
`Journals ABMnrg
`
`MINIHEUIEW
`
`Pharmacokinetic and Pharmacodynamic Considerations in
`Antimalarial Dose Optimization
`
`Nicholas J. White
`
` I"-.-'lj|'..-'_!»:I '-,-.~:'-_!
`:-'-.—:-:,-_-'1|-;|'. -.i';-I_.F1.;|. |_‘-.-' -:| "Ic-L- -:':|
`-::..—'_-
`
`
`
`"-.'.-.l'-:_|r-:i '_=r|
`-_
`-.-
`'. t|.| I?-'2l
`l-..n.__.-._. .. .'
`
`.‘-.-1:--;--_'I'-~..
`
`."'.'l_l.'.|'_'l-_'l
`
`.I'
`
`|-.r—_u
`
`1.1"._.I -'.-| .TI'-;-|3r:-:'--Ze!'u€
`
`"on Ti-:-i_-u;;:| i-.-'l-:r.‘l;:r--+,I:lu...-_'nli I :;-:-::.I_.
`
`Antimalarial drugs have usually been first deployed in areas ofmalaria endemicity at doses which were too low, particularly for
`high—risk groups such as young children and pregnant women. This may accelerate the emergence and spread of resistance,
`thereby shortening the useful life of the drug, but it is an inevitable consequence o fthe current imprecise method ofdose fi nd—
`ing. An alternatiVe approach to dose finding is suggested in which phase 2 studies concentrate initially on pharmacoldnetic—
`pharmacodynamic (PKHPD) characterization and in viva calibration of in vitro susceptibility information. PD assessment. is
`facilitated in malaria because serial parasite densities are readily assessed by microscopy, and at lowr densities by quantitative
`PCR, so that initial therapeutic responses can be quantitated accurately. If the in viva MIC could be characterized early in phase
`2 studies, it would provide a sound basis for the choice of dose in all target populations in subsequent combination treatments.
`Population PK assessments in phase 213 and phase 3 studies which characterize PK differences between different age groups, clin-
`ical disease states, and human populations can then be combined with the PK—PD observations to provide a sound evidence base
`for dose recommendations in different target groups.
`
`he primary objective of treating severe malaria is to save life.
`Other considerations such as preventing recrudescence or mi—
`nor toxicity are secondary. 1n uncomplicated malaria. the main
`objective of antimalarial drug treatment is cure of the infection.
`Speed of response is also important. as this reflects the rate at
`which the disease is controlled and the corresponding reduction
`in the risk of progression to severe malaria. Lesa—Serious adVerSe
`effects therefore become a more important factor in determining
`dose. The therapeutic response in malaria is determined by the
`concentration profile (pharmacokinetics Phil) of active antima—
`larial drug or drugs in the blood (as the asexual parasites which
`cause malaria pathology are confined to the blood). tlieirintrinsic
`pharmacodynamic {PD} properties, the susceptibility of the in—
`fecting parasites to the drugls), the number of asexual malaria
`parasites in the blood, and the activity of host-defense mecha-
`nisms. Ideally, antimalarial treatment should be 100% effective in
`everyone, but this may not be possible without producing toxicity
`or recommending a long course of treatment with consequent
`poor adherence.
`[t is now recommended that all antimalarial
`treatments for uncomplicated malaria should aim at :1 >95% cure
`rate for the blood-stage infection it). In recent years. a general
`agreement has been reached on methods ofclinical and parasito—
`logical assessment to measure the cure rates in casas of uncompli~
`cared falciparum malaria (L—3). ln Plasmodinm virus: and P. [ii-vile
`infections, persistent
`liver—stage parasites (hypnozoitesj cause
`later relapses, despite cure of the blood~stage infection, which
`complicates therapeutic assessment. These infections require ad—
`ditional treatment with Seaminoquinolines [radical cure). Re-
`lapses are often genetically heterologous and cannot be distin—
`guished reliably from recrudescen ces or new infections. This
`necessitates a different approach for assessment of treatment Elfi—
`cacy in the relapsing malariasfiwhicli is yet to be agreed upon.
`Many ofthe antimalarial drugs in current use were introduced
`at suboptimal doses. For various reasons, quinine, sulfadoxine—
`pyrimethamine, primaquine {for radical cure oftropical frequent
`relapsing P. vivax infections). mefloquine. halofantrine. artemis-
`
`inin derivatives, artemether-lumefantrine, and dihydroartemis—
`inin—piperaquine (i.e., 7 of the 12 current antimalarials) were all
`deployed initially at doses which were too low in some or all age
`groups. Pyrimethamine and sulfadoxine (310535 for children were
`extrapolated from experience in Caucasian and Asian adults.
`Their pharmacohineticproperties Were not studied in younger age
`groups before widespread deployment in Africa, where children
`are the main target group [4). The dose was too low in young
`children. The primaquine dose regimen (15 mg baseiday adult
`dose} was developed largely on the basis of studies of the long—
`latency Korean vivax malaria, but this close was then recom—
`mended widely in areas with the more resistant tropical relapse P.
`vivax phenotypes (5). 111 Southeast Asia and Oceania, this dose is
`too low. Five—day primaquine regimens were deployed very widely
`for radical cure ofvivax malaria for ovsr 30 years—yet these reg—
`imens were largely ineffective. Fourteen—day courses are now rec—
`ommended. Mefloquine was first introduced at a single dose of] 5
`mg baselkg of body weight (-6, 7}. which may have hastened the
`emergence ofresistance (-8}. The total dose now recommended is
`25 mgikg divided over 2 or 3 days. The closes ofartemisinin deriv—
`atives used initially as monotherapy, and then subsequently in
`combination treatments {arteniether at 1.6 mgikgidose in arte—
`mether—lumefantrine and dihydroartemisinin at 2.5 mgikgidose
`togetherwith piperaquine}, may not provide maximal effectsin all
`patients. The initial treatment regimen of artemether—lumefan—
`trine deployed was a four—dose regimen which provided insuffi—
`cient lumefantrine and gave high failure rates (six doses are now
`recommended} {'9'}. The dose of dihydroartemisinin in the first
`
`«xi-lemon '_:I"'
`Published ahead of paint E-
`
`dd .1 It: -.
`urn.reins-5.5..
`'.-.._ II-_\|.E.
`.
`'il'i’l .IL'L'. n... r
`-.r_--_-r_1r-_-r.;_-:
`
`.-_all
`r .-1.ng ,a.;r -_ _., L
`
`|-_'|_‘|‘I--j.t1|lj‘|1:|jIjr..iy[r|.—
`_..,l-|'
`. Filth? I'l-|_
`If: I_I[.'i‘"lI-_"
`
`'-| Ir“:- -'_|>-'.=|t:vt-'-i_'- Whit-.2”: Atir'!'llitu.~l|
`3-
`'“Ill' tie»! ||-. LIE-"-
`|rv|'n-'
`.«.-.-1|'- i
`l _"jfr\r’tr'
`in."
`
`5192 sarcasmiorg
`
`Antimicrobial Agents and Chemotherapy
`
`p. 51392—580?
`
`December 2013 l.l'olume- 5? Number i2
`
`
`
`
`
`isonfiliq“oz“9|,Menueruoifiiowseoeeyzduuwonpapemumog
`
`InnoPharma Exhibit 10300001
`
`

`

`Minireview
`
`formulations of the diltydroartemisinin—piperaquine combina—
`tion was <2 mgr‘kg (it is now 2.5 mg/kg, which may still be too
`low) {10-}. The pharniacokinetic properties ofpiperaquine are dif-
`ferent in children from in adults, and there is evidence that current
`
`dosing schedules in children may be suboptimal (l 1). After 3 cen-
`turies of reasonable dosing based originally upon the Schedule
`Romana. treatment recommendations for quinine in severe ma—
`laria were suddenly reduced in the l970s to a doSe as low as 5
`mgr‘kgiltl h. which is eight times lower than that now recom—
`mended. In contrast. the quinine loading dose in severe malaria
`was not introduced until the early 19805. and it is still not recom—
`mended universally (.12). The initial recommendation for artesu—
`nate treatment in severe cases ofrnalaria was a daily maintenance
`dose ofhalfthe initial dose (1.2 mgikg}. As oral bioavailability is
`approximately 60%. this corresponds to an oral dose of 2 mglkg
`{1-3, 14}. The currently recommended parenteral close is twice this
`and is the same as the recommended initial dose, 2.4 mgl'kgr‘day
`(1'. 15. 1th. Recent evidence suggests that this dose should be in—
`creased in young children (1'7).
`Optimizing drug dosing requires characterization ofthe phar—
`niacokinetic and pharmacodynamic properties of the drug in the
`target populations. There are four main determinants of the ther—
`apeutic response: antimalarial pharniacokinetics {affected byvari-
`ables such as coadministration with food. age, pregnancy, disease
`severity, vital organ dysfunction, partner drug, and coinfectionsi
`other drugs), parasite susceptibility {incorporating effects on dif—
`ferent stages of asexual parasite development. dormancy, propen-
`sity for resistance to develop, and level of resistan ce first selected],
`host defense (influenced by age, pregnancy, and transmission in-
`tensityiexposure history}. and parasite burden. In addition. mixed
`infections can be a factor. For antimalarials. ex vivo systems are
`useful for predicting resistance (18] and they provide valuable
`pharmacodynamic information (19}, but they are simply not
`good enough yet to replace in vivo evaluations for dose finding. In
`uncomplicated falciparum malaria,
`it
`is generally agreed that
`combinations, preferably,
`fixed—dose
`combinations
`{FDCL
`should be used. The same should apply to vivax malaria. although
`chloroquine and primaquine can be considered a combination.
`When drugs are first developed, there is a limited window of op—
`portunity to define the dose—response (or concentrationveffect)
`relationship for the single new compound. but this opportunity
`must be taken {20). Once the drug is available only as an FDC, the
`dose ratio is. by definition. fixed and it is too late for optimization
`ofthe individual component doses. Characterizing the individual
`drug dose— response relationships is essential for rational dose op—
`timization, and so a good drug development approach involves
`documenting the blood concentrations that are associated with
`submaitimal antimalarial effects. Studies in animal models. partic—
`ularly with P. jitlciparnm, may be informative. but studies in hu-
`mans will also be needed. It is important to accept that this may
`result in temporary therapeutic failures in some vol unteers. There
`is a natural reluctance to accept this. but sensitive detection meth—
`ods to measure low parasite densities now provide us with safe
`methods that should avoid any risk or discomfort to the patient
`(2]). Suggestions are provided here for an alternative PK-PD ap-
`proach for dose finding which, ifvalidated, may improve and ac—
`celerate dose finding and so avoid systematic underprescribing
`and thus underdosing. It might also prove more rapid and less
`expensive. The primary objective is determination of the in viva
`MIC as the basis for rational dosing (the MIC is the concentration
`
`Total
`
`Detection limit
`
`parasites
`
`
`
`tsenfianArea“9|,MenueruorfiJo'queoeeyzduuwonpepemumog
`
`Weeks
`
`FIG I Population PK PD responses lollowinga 3 -day Ircatmcnl will: a hypothet-
`ic al slowly eliminated :ml'irn alarial drug. The total numbers ofmalaria parasites in
`the body over time are depicted in blue in a range of patients presenting with
`parasile densities between approximately 50 and ZULDUOHLL The ranges ofdrilg
`concenlration profiles are shown in red. with the corresponding ranges of parasi-
`tological responses in blue. Parasilclnia levels cannot be counted reliably by Ini
`croscopy below 50nd [Corresponding to ' 100,000,000 parasites in the body ol'an
`adnll}. The M'PC is the lowest lslrmd,plasma1 or free plasma col Icel'ltrnlion which
`produces Ille Inzuilnulrl parasllicitlal eilecl {Len Ihe maximum parasite reduction
`n1hr)l.‘lhis corresponds lo the concentration associated wilh first slowing of the
`first order (log linear) decline in panniternia.
`
`at which the parasite multiplication factor per asexual cycle is 1}. It
`is necessary first to consider the factors which affect the pharma—
`cokinetic properties of antim alarials in malaria and then to con—
`sider antimalarial pharmacodynamics and how PK—PD relation—
`ships should be assessed.
`
`PHARMACOKINETICS
`
`The pharrnacokinetic (PK) properties of antimalarial drugs are
`often altered in patients with malaria compared with healthy sub—
`jects. The PK properties therefore change as the patient recovers.
`PK properties are also often significantly different in important
`patient subgroups such as young children and pregnant women
`{22). Several of the antimalarial drugs, notably those which are
`hydrophobic and lipophih'r.1 are poorly absorbed after oral or in—
`tram uscular administration and Show wide interindividual differ—
`
`ences in concentration profiles. In general. this variation in blood
`concentrations is inversely proportional to bioavailabiljty, which
`emphasizes the importance of improving bioavailability in drug
`development. Increasing bioavailability provides the twin benefits
`of reducing the required dose and thus the cost ofthe drugs and
`reducing the individual probabilities of underdosing or overdos—
`ing. In considering antimalarial dosing in the past, we tended to
`concentrate on mean or median values ofPK variables, but itis the
`
`patients with the lowest blood concentrations who are most likely
`to fail treatment and facilitate the emergence of resistance and
`those with the highest concentrations who are most likely to ex—
`perience drug toxicity (23). These extremes need to be defined.
`which means that characterizing the distributions of PK variables
`in important target groups is as important as assessing their mea—
`sures of central tendency (Fig. 1). Characterizing these distribu—
`tions well eventually requires sampling of relatively large numbers
`ofpatients, which in turn usually necessitates sparse sampling and
`population PK modeling. Optimal design approaches can be used
`to ensure that the information is gathered most efficiently (24). It
`
`December 2013 Volume 5? Number ‘IZ
`
`aacasmmg 5793
`
`lnnoPharma Exhibit 1030.0002
`
`

`

`Minireview
`
`is essential that key patient groups such as young children and
`pregnant women are studied specifically, and there should be a
`postregistration commitment to this if such investigations have
`not been conducted during preregistration studies. There may
`also be clinically relevant pharmacogenetic differences in drug
`metabolism between different ethnicgroups. Thus. characterizing
`the distributions ofpharmacokinetic variables is a gradual process
`accrued during phase 2 and phase 3 of drug development, but it
`must continue into phase 4 to cover all relevant populations.
`Malaria is often worst in remote rural areas. The recent devel—
`
`opment ofsimple methodologies such as drug measurement from
`capillary blood filter paper samples (25. 26) will facilitate commu-
`nity—based assessments in remote settings and make sampling of
`infants and children feasible. Thus, population PK information
`will eventually be needed in all important target groups (i.e.. in—
`fants, children, pregnant women. lactating women, malnourished
`patients, patients receiving antituberculosis lanti—TB] and antiret—
`roviral drugs. etc.) {22) to provide optimal dose recommenda—
`tions. There is currently limited bioanalytical capacity to support
`such studies. but there are international schemes to assist antima—
`
`larial drug measurement and ensure the accuracy of the results,
`which should facilitate future laboratory bioanalytical capacity
`development in tropical countries (27. 28}.
`In drug development. where a new compound has not been
`used previously, there is little information on distributions of PK
`variables and so the important but difficult issue is to determine
`how much PK—PD information is enough to decide upon a dosage
`recomm end ation. For safety reasons, the PK information is usu-
`ally gathered in the following standard sequence: experimental
`animals, healthy normal volunteers, adult patients with uncom—
`plicated malaria. children, and, much later, infants and pregnant
`women.
`
`PHARMACODYNAMICS
`
`(i) Action ofthedrugs. The antimalarial drugs differ in their stage
`specificities of action against malaria parasites. The S—amino—
`quinolines are unusual in killing pre—erythrocy‘tic—stage parasites,
`hypnozoites. and mature ganietocytes ofP.fi.ilciparnm but having
`weak activity against its asexual stages (2-9). They are more active
`against asexual stages of P. vivax and P. lcuuwlesi. All other antiv
`malarial drugs in current use kill the asexual and sexual stages of
`sensitive P. Vii-'(LY, P. mnlnrinc, P. ovals, and P. knowlesi and the
`asexual stages and early gametocytes (stages 1 to [1]) ofsensitive P.
`julcrparnm. but they do not kill the mature P. jitltiipumm gameto-
`cytes (stage V). The artemisinins have a broader range ofeffect on
`developing P. jiilciparmn sexual stages, as they also kill stage IV
`and younger stage Vgametocytes. Atovaquone and the antifols kill
`preerythrocytic stages and have sporontocidal activity in the mos—
`quito {interfering with oocyst formation and therefore blocking
`transmission). Apart from the 8—aminoquinolines, none of these
`drugs have significant effects on P. vivax or P. made hypnozoites.
`Even within the asexual cycle there are differences in antimalarial
`activity in relation to parasite development. None ofthe currently
`used drugs have significant effects on very young ring stages or
`mature schizonts. and all have their greatest effects on mature
`trophozoites in the middle of the asexual cycle (36). In addition,
`the artemisinins (and other antimalarial peroxides) have substan-
`tial ring—stage activity which underlies their life—saving benefit in
`treatment ofsevere falciparum malaria (15, 16. 31}. Several anti-
`malarials. notably. some antibiotics with antimalarial activity.
`
`have greater effects in the second than in the first drug—exposed
`asexual cycle (23]. The pharmacokinetic—pharmacodynamic rela—
`tionships (PK—PD) have not been very well characterized for any
`ofthese activities.
`
`(ii) In vivo pharmacodynamic measures. In severe malaria.
`the primary therapeutic concern is the speed of parasite killing
`and, in particular. the killing of circulating ring—stage parasites
`before they mature and sequester (39, 3] ). Rapid killing ofyoung
`P. jiilcipurrrrn parasites by artemisinin and its derivatives explains
`much of the superiority of artesunate over quinine in the treat~
`ment ofsevere falciparum malaria [15, 16). in uncomplicated ma—
`laria, rapid ring—form killing is also important, as it contributes to
`the speed ofpatient recovery, but the main therapeutic objective is
`to reduce para site multiplication. Once iurtinlala rial treatment is
`started. then. after a variable lag phase, parasite killing in vivo
`approximates to a first-order process {32-34) as represented by
`the following equation:
`
`P, — Pue' tr
`
`(1 i
`
`where P[ is the parasitemia level at any time I after starting treat—
`ment, PD is the parasitemia level immediately before starting treat—
`ment. and kg, is the first—order parasite elimination rate constant.
`The parasite clearance half- life is therefore 0.693hl‘r. In equation I.
`parasite killing equates with parasite removalfrom the circulation.
`but in falciparum malaria (but not the other malarias} there is an
`additional major factor removing parasites from the circulation.
`and that is cytoadherence. Only parasites in the first third of the
`asexual cycle circulate, and the more mature parasites are seques—
`tered. This complicates interpretation of the parasite clearance
`curve following treatment with drugs which do not kill ring-form
`parasites, as initial declines in parasitemia result mainly from se—
`questration and not drug effects (33). Parasite killing can be ex—
`pressed as the parasite reduction ratio (PER). which is the frac—
`tional reduction in parasite numbers per asexual cycle. or the
`reciprocal of ring-form k1,. per cycle {32). This cancels out the
`effects ofcytoadherence, as the parasite populations are assessed at
`the same stages ofdeveloprnent separated by one cycle. The shape
`ofthe concentration~effect relationship in viva is assumed always
`to be sigmoid. as it is in vitro (Fig. 2), per the following equation:
`
`k _ kin-1.x" [Cn/ECSHH l {:nl
`
`where k is the parasite killing rate and ileum is the maximum par—
`asite killing rate (i.e., the maximum effect, orljnm) for that drug in
`that infection, C is the concentration of drug in blood or plasma.
`ECE,J is the blood or plasma concentration resulting in 50% of the
`maximum effect, and n is a parameter defining the steepness ofthe
`dose—response relationship. For most drugs, maximum effects are
`probably achieved initially. The evidence for this is the lack of a
`relationship between peak concentrations and parasite clearance
`(the exception is quinine treatment of severe malaria without a
`loading dose, which provides submaximal effects in some pa~
`tients} (1'2). So while concentrations exceed the minimum para—
`siticidal concentration {MPC).lc in equation I is equal to kum. It
`should be noted that each end ofthe sigmoid curve approaches 0%
`and 100% effects asymptotically—“so the MPC is an approxima—
`tion, whereas the ECSU is a more robust and precise estimate. Once
`antimalarial concentrations in blood decline to a level below the
`
`MPC. the parasite killing rate declines (see the “Antimalarial phar—
`macokinetic—pharmacodynamic relationships" section below).
`For drugs in current use, maximum PRRs range from approxi—
`
`5794 aacasrnorg
`
`Antimicrobial Agents and Chemotherapy
`
`
`
`isenfisq“oz“9|,Menueruo{file'uiseoeemduuwonpepemumog
`
`lnnoPharma Exhibit 1030.0003
`
`

`

`
`
`Log concentration
`
`FIG 2 The concentration cll'ect relationship; for antimalarial drugs, the effect
`is parasite killing, which can be measured in different ways. The Ema); is the
`maximum parasite killing Iliat a drug can produce, which translates in viva into
`the maximum parasite reduction ratio. The litjffl, is the blood or plasma con
`cent ration providing 50% of matimum killing. The median and range values
`for a hypothetical population ofmalaria parasites are shown in blue. and lllc
`dislribulinu of average drug levels in palieuts is shown as :1 Yeti bell-shaped
`curve ( i.e., concentrations are log normally distributed}. Clearly, some of the
`patients have average drug levels below the MPG and would not have maxi
`mum responscswilh this dose regimen.
`
`mately 10—fold to approximately 10,000—fold reductions in para—
`sitemia per asexual cycle. The mean values and their variance in
`vivo have not been established for several important antimalarial
`drugs in current use (notably lumefantrine and piperaquine}. and
`for others, where monotherapies have been evaluated. the esti—
`mates are often imprecise. There is no evidence for saturation of
`parasite clearance, but, obviously, the higher the initial bio—
`mass, the longer it takes to eliminate all the parasites from the
`body (3-3). Consequently, patients with high—biomass infec—
`tions need more antimalarial drug exposure than those with
`low—biomass infections.
`
`(ii) In vitro susceptibility. For antimalarial effects, the shape
`and position ofthe concentration-effect curve studied ex viva de-
`pends on the susceptibility of the infecting parasites and the PD
`readout (typically, for blood stages. inhibition ofgrowth or mat—
`uration, inhibition of hypoxanthine uptake. inhibition ofprotein
`or nucleic acid synthesis, etc). Furthermore, each in vitro method
`assesses a slightly different section ofthe asexual life cycle, which
`may result in important differences between methods in the re-
`suits for drugs with ring—stage activity. It is not clear exactly how
`the effects ofthese static drug concentrations in a small volume of
`dilute blood in the laboratory correspond with in vivo effects (1-8,
`19, 35). Neither is the relationship between inhibition of parasite
`growth and subsequent inhibition of multiplication well estab~
`Iished. inhibition of growth is measured in most in Vin-o tests,
`whereas in in viva patient studies, inhibition of multiplication
`( parasite clearance} is recorded. In the absence of in viva i nforma—
`tion on the concentration—effect relationship. for predictive mod~
`cling purposes the slopes ofthe linear segments of the in vitro and
`in vim sigmoid concentration-effect relationships have been as—
`sumed to be similar (8, 35), but whether or not such an assump—
`tion is justified remains to be established. Most agree that the
`antimalarial drug concentration that is biologically relevant in as—
`sessilig blood~stage effects is the (unbound) fraction in plasma.
`Total red cell concentrations are less informative as the parasitized
`
`Minirev'iew
`
`red cells behave very differently from their unparasitized counter-
`parts. In the patient, the blood concentrations of the antimalarial
`drug are changing constantly, and the parasite age distributions
`may differ considerably between patients. Ex vivo systems with
`changing antimalarial concentrations that are more biologically
`relevant than the simple static drug susceptibility assays have
`therefore been developed, and measurement ofmultiplication in—
`hibition can yield valuable information (19}. Rodent models
`capable of sustaining human malaria infections have also been
`developed recently {56). Human malaria infections in irnmuno—
`deficient mice allow PK—PD characterization and thus provide
`useful information in predicting therape otic responses in patients.
`These laboratory studies have the great advantage that parasites
`from many different locations or with known resistance profiles
`can be studied and compared. it is argued below that if the rela—
`tionship between the standard in Vin-n susceptibility measures
`{50% inhibitory concentrations [[CSOI,
`lCaU, etc.) and in vivo
`PK—PD responses in patients with malaria could be characterized.
`then this would facilitate dose finding.
`
`ANTIMALARIAL PHARMACOKINETIC-PHARMACODYNAMIC
`RELATIONSHIPS
`
`Some of the best research on antimalarial PK-PD relationships
`came from the period ofintense antimalarial drug investigation in
`the United States during and shortly after the Second World War
`(Fig. 3]. Studies were conducted to determine the optimum dos—
`ing strategies for mepacrine (atebrine, quinacrine), the Cinchona
`alkaloids, and both the 4— and S—arninoquinolines (37—39). Phar—
`macokinetic analysis had yet to he invented, and methods for
`quantitation ot‘clrugs in serum or plasma were in their infancy. but
`the spectrophotometric assays that were conducted still provided
`valuable information. Relatively large numbers of nonimmune
`adult male volunteers artificially infected with single "strains" of
`P. faldpnrum or P. vivnx received different dose regimens. serum
`levels were measured, and therapeutic responses were assessed.
`This research provided dose~response or concentration-effect re~
`lationships and led to the mepacrine loading—dose regimen. char—
`acterization of the comparative antimalarial effects of the four
`main Cinchona alkaloids (quinine, quinidine, cinchonine, and.
`cinchonidine), and development of the standard dosing regimen
`for chloroquine [one ofthe few antimalarial dose recommenda—
`tions which has stood the test of time). This was still the era of
`malaria therapy, and the war had focused military attention on
`malaria. Such volunteer studies are no longer possible today. Since
`that time, PK— PD relationships have been inferred mainly from
`clinical studies ofantimalarial treatment (8, 9, 4043).
`(i) PIC-PD correlates. Studies ofPK—PD relationships for anti—
`bacterial effects have shown that for some antibiotics (those with
`steep concentration-effect relationships and without postantibi~
`otic effects), bacterial killing is dependent on the duration for
`which the antibiotic exceeds the MIC for the bacterial population
`(“time above MIC”). For other antibiotics (where concentrations
`achieved with current regimens remain on the steep part ot'the
`concentration—effect relationship), it is the maximum concentra—
`tion achieved (Cum) or the related area under the plasma concen—
`tration—‘cime curve (AUC) that is the best correlate of bacterial
`killing (Fig. 4). These PK variables are all interrelated lie, the
`higher the Cm“, the larger the AU C and the longer the time above
`the MIC]. With some adjustments, these PK measures can be ap—
`plied to antimalarial effects {32). although correlates with parasite
`
`
`
`
`
`isenfiitq“oz‘gLMenueruorfiiouiseoeenzduuwonpepemumog
`
`December 2013 Volume 5? Number ‘IZ
`
`aacasmmg 5795
`
`InnoPharma Exhibit 1030.0004
`
`

`

`Minireview
`
`
`
`Plasma
`
`+ Class]
`
`0 Class [I
`
`o Classlll
`
`quininemgfl.
`
`0.1
`
`0.2
`
`0.3
`
`0.4
`
`0.5
`
`Daily dose {Grams}
`
`Effect
`
`
`
`
`
`isenfiso1m:‘5”,Manneruo{Browseoeemduuwonpepemumog
`
`2 — Gill-11910310 -
`I
`I
`l
`0
`I
`
`ll
`
`O
`
`5
`
`'lO
`
`Plasma concentration {mglL}
`
`FIG 3 Dose—response relationships oblainecl between [he years [915 and 1946 for quinine in blood-induced Vivax malaria {McCoy strain} in volunteers (33).
`Plasma concentrations after protein precipiialion were measured spectropliotomelricadly, which overestimales parenl compound concentralions. The left box
`shows the variable relationships between dose and mean plasma concentral'ituis. and the right graph shows the concentration effect relatioi'lsliip divided into
`three effect measures: class 1. no certain effect; class i], temporary suppression of parasitemia audior fever: class III, “permanent” effect. i.e.. absence ol
`parasitenlia for M days.
`
`killing have not been established for most antimalarial drugs.
`Whereas rnost bacteria replicate every 20 to 40 min, asexual ma~
`lari'a parasites infecting humans replicate every I to 3 days. Symp—
`tomatic infections usually comprise one predominant brood of
`malaria parasites, but multiple genotypes are often present—par-
`ticularly in higher—transmission settings—and so within one host
`there may be subpopulations with different drug susceptibilities
`(and also different stages of asexual development). The lowest
`blood. plasma, or free plasma concentration which produces the
`maximum PRRis the MPC (Fig. 5}. These PK—PD variables reflect
`the antiparasitic effects of the antimalarial drug and host immu-
`nity and so are specific for an individual and that individual’s
`
`AUGMIC
`
`Concentration
`
`Weeks
`
`FIG 4 Plasma or blood concentration profile of a slowly eliminated aritirna
`larial drug showing an arbitrary MIC. The AUC is the area under theciirVe,ancl
`Ctnax is Ihe maximum concenlmtion in blood or plasma. AUC from 7 days to
`infinity is shown in darker pink. Blood concentrations are increasingly mea-
`sured on day Y in therapeutic assessments of slowly eliminated antimalarials
`{49).
`
`infection. Innate host—defense mechanisms and acquired imm Line
`responses contribute significantly to therapeutic responses—ef—
`fectively shifting dose—response curves to the left. The contribu—
`tion of the host immune response, which may be significant even
`in previously no uimm une patients 44). has not been well char—
`acterized.
`
`With current dosing for all antimalarial drugs except the cute
`misinin derivatives. drug elimination is sufficiently slow that the
`antimalarial eliects ofa treatment persist for longer than one asex—
`
`
`
`Totalparasites é.
`
`Detection Ilrnll'.
`
`
`
`
`Weeks
`
`FIG 5 Different therapeutic re'stISes to a slowly eliminated anlill‘lalari‘al
`ilmg in a malaria infection of mm parasites {parasite density,
`-2,000l|.|.l). The
`blood concentration profile in gray is shown in the background. Parasitologi
`cal responses range from fully sensitive {green} to highly resistant. {blue}. Each
`response is associated wil h :1 dil-l‘erenl level of snscep Iibili ty and lhns a dillerenl
`MIC and MPC [arrows polnlillg to collceniralion profile}. The inset repre-
`sents the concentration effect relationship {or Lllc lowest level of resistance
`(resulting in 11 Ian: failure} showing correapoi‘lding points for the MIC and
`MPC (orange curve}.
`
`5796 aacasmorg
`
`Antimicrobial Agents and Chemotherapy
`
`
`
`‘.
`Ia
`
`Jiat
`
`.'1
`t
`r
`
`lIlllll-1‘.II.Il‘1111I11
`i111111i1I111i11iiIiIi1111
`
`lnnoPharma Exhibit 1030.0005
`
`

`

`Minireview
`
`ual cycle (8, 22. 32. 45). Indeed, many antimalarials have terminal
`elimination half—lives (t, mid) of several days or weeks. in order to
`cure the blood-stage infection in a nonimmune patient, antima—
`larial concentrations in blood {free plasma concentrations] must
`exceed the MIC for the infecting parasites until the last parasite is
`killed (8, 22, 23. .32. 45). The higher the initial parasite burden. the
`longer this takes. With host immunity. cure of malaria may be
`achieved even it'drug levels fall below the MIC before complete
`elimination ofall parasites {44). Thus, the time above the MIC is
`an important PK determinant oftherapeutic outcome, although
`the AUC above the MIC is also relevant. as the rate of parasite
`killing is determined by the concentratiomeffect relationship
`above the MIC for the infecting parasites and by the antimalarial
`concentration profile in the treated patient.
`Assuming that the parasites are exposed to the antimalarial
`drug at a sensitive stage, what duration of exposure in a single
`asexual life cycle is necessary for maximum effect? For sensitive
`parasites. it appears that up to 4 h of exposure is required [31),
`although for some drugs less time is needed. For artemisinin de

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