throbber

`
`AAC
`Jo-.J'nals F\'.:iil.I'l.L1lg
`
`MINIHEUIEW
`
`Pharmacokinetic and Pharmacodynamic Considerations in
`Antimalarial Dose Optimization
`
`Nicholas J. White
`
`I"-.-"l 3|'..-Ll»: I '.-.~_--_! u;:..-'_ :7-.—:-:.—_-an.-;|'. -_i';-n_.F,.;u.
`
`".-.-.l-:_:r-:i'_=r:
`-_
`.:
`_
`..
`'.ru.Ic-'1 l-..r|-._.-._....'
`
`|_‘-.-' -:|
`
`'|-,'-I.- -lsl .‘-.-1:-lg.-_'ru~_.
`
`."-.]_1.'.:-_'I._u|
`
`,|'
`
`:-..r—_-:
`
`-'__1I'._.I
`
`-'.-I .TI"-5-Iar:-:"'Z-%:'{I% "cu Ti-I't_':I1.':l J-.-'l-:r;n;::'~+,CIu...-_'nIi I ;;u:::.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 usefi.I.l life of the drug, but it is an inevitable consequence o fthe current imprecise method ofdose ii ud-
`ing. An alternative approach to dose finding is suggested in which phase 2 studies concentrate initially on phat-macokinetic—
`pharmacodynamic (PK~PD) characterization and in viva calibration of in vitra susceptibility information. PD assessment. is
`facilitated in malaria because serial parasite densities are readily assessed by microscopy, and at low 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 2b 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. In 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. Less—serious adverse
`effects therefore become a more important factor in determining
`dose. The therapeutic response in malaria is determined by the
`concentration profile (pharmacokinetics lPi=il_} of active antima-
`larial drug or drugs in the blood (as the asexual parasites which
`cause malaria pathology are confined to the blood). then-intrinsic
`pharmacodynamic {PD} properties, the susceptibility of the in-
`fecting parasites to the drugfsl, the number of asexual malaria
`parasites in the blood, and the activity of host-defense mecha-
`nisms. ldeally. 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 adlierence.
`It is now recommended that all antirnalarial
`treatments for uncomplicated malaria should aim at 21 .“>95% cure
`rate for the blood-stage infection (1). In recent years. a general
`agreement has been reached on methods ofclinical and parasito—
`logical assessment to measure the cure rates in cases of uncompli-
`cated falciparurn m:-ilaria (L-3). ln Plnsmodinm 1u'vu.\: and P. [I1-‘tilt?
`infections, persistent
`liver—stage parasites fhypnozoitesj cause
`later relapses, despite cure of the blood~stage infection, which
`complicates therapeutic assessment. These infections require ad-
`ditional treatment with 8—aminoquinoline5 [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 eth-
`cacy in the relapsing rnalarias-wln'ch 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. v1'vnxinfections). mefloquine. halofantrine. artetnis-
`
`inin derivatives, artemether—lumefautrine, 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 sulfadoitine doses for children were
`extrapolated from experience in Caucasian and Asian adults.
`Their pharrnacoltineticproperties 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 basefday adult
`dose} was developed largely on the basis of studies of the long-
`latency Korean vivax rnalaria. but this dose was then recom-
`mended widely in areas with the more resistant tropical relapse P.
`vfvnx phenotypes (5). in Southeast Asia and Oceania, this dose is
`too low. Five—day primaquine regimens were deployed very widely
`for radical cure ofvivax malaria for over 30 ye-ars—yet these reg-
`imens were largely ineffective. Fourteetrday courses are now rec-
`ommended. Mefloquine was first introduced at a single dose of] 5
`mg baseikg of body weight (-6, 7}. which may have hastened the
`emergence ofresistance (-8). The total dose now recommended is
`25 mgfkg divided over 2 or 3 days. The doses ofartemisinin deriv-
`atives used iuitially as monotherapy. and then subsequently in
`combination treatments (artemether at 1.6 mgfkgfdose in arte-
`rr1ether—lu1nefantrine and dihydroartemisinin at 2.5 rngfkg/‘dose
`togetherwith piperaquine}, may not provide rnaximal effectsirt all
`patients. The initial treatment regimen ofartemether—lun1efan—
`trine deployed was a four—dose regimen which provided insuffi-
`cient lL1mefant1'ine and gave high failure rates (six doses are now
`recommended) ('9). The dose of dihydroartemisinin in the first
`
`'_::n'-
`1._-[.I,;-r:._',.,'_|
`Published ahead of pn'nr E-
`»\.d _1Ir_-.
`-.
`-.-,:.-_.- I_:j-|junu:-._1I:_-'_...;._
`-,.'y'|.I1.'i_-_ "l'_ .-
`'.-:._ u._.|_3_ _
`'_ |'_--_-r_1'-_-n:_-:
`"_-_-
`
`|-_':_-.1.-;.tI|r_.:1:.j I][ul:‘[ 1|.-
`._-‘-.i|u.'v_e
`ii.._
`_-_o||ru'1.fr|,'«.:r_:__.l._
`
`
`
`
`
`.~.-.-1.- ii _'~:!A#.r' NI_-."
`
`
`
`
`
`isanfiliq,i_|,oz‘gtManneruo,rfSio'uJse'oee;;:d1J,uwon|‘.'J9pE0|UMOG
`
`5?92 aacssn1.org
`
`Antimicrobial hgents and Chemotherapy
`
`p. S?92—58O.7
`
`December 2013 Volume 5? Number 12
`
`InnoPharma Exhibit ‘lO30.000‘l
`
`

`

`Minireview
`
`formulations of the dihydroartemisinin-piperaquine combina-
`tion was <2 rnglkg (it is now 2.5 mg/kg, which may still be too
`low) [10]. The pharmacokinetic 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 Schedula
`Roniana. treatment recommendations for quinine in severe ma—
`laria were suddenly reduced in the 19705 to a dose as low as 5
`niglkg,f.‘»'_4 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 1980s. and it is still not recom—
`mended universally (.12). The initial recommendation for artesu—
`nate treatment in severe cases ofmalaria was a daily maintenance
`dose ofltalfrhe initial dose (1.2 mgfkg}. 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 mglkglday
`(I. 15.
`lei}. Recent evidence suggests that this dose should be in-
`creased in young children {l'7').
`Optimizing drug dosing requires characterization ofthe phar-
`niacokinetic and phartnacodyn-antic 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 coinfections!
`other drugs), parasite susceptibility {incorporating effects on dif-
`ferent stages of asexual parasite development. dormancy, propen-
`sity for resistance to develop, and level ofresistan ce first selected],
`host defense (ilifluenced by age, pregnancy, and transmission in-
`tensitylexpostire history}. and parasite burden. In addition. mixed
`infections can be a Factor. For antimalarials. ex viva 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 viva 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 prirnaquine can be considered a combination.
`When drugs are first developed. there is -a limited window of op-
`portunity to define the dosevresponse (or concentration—effect__l
`relationship for the single new compound. but this opportunity
`must be taken (20). On ce 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
`5I.tl3l'l"lI-J..V{‘lt'I‘tal antimalarial effects. Studies in animal models. partic-
`ularly with P. jiilcipamrm, may be inforrnative. 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
`(21). 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
`
`
`
`
`
`isanfiliq,i_|,oz‘gtManneruo,rf3.Io'uJse'oea;,r:diiumoi;pepeolumog
`
`Weeks
`
`FIG I Poplllation PK -PD responses lollowinga 3 —day Ircannenl with a l|ypoll1el—
`l( :1] slowly eliniinatvd a.ril'i1'n alarial drug. The total numbers olinalaria ptuasitcs in
`the body over time are depicted in blue in it range of patients presenting with
`parasite densities between apprrtcitruately 50 and 20U,000.",ul. The ranges nltlrug
`concentration profiles are sliowll in red. with the corresponding ranges {ill p-.1ra.si-
`tological rcsportscs in blue. Parasitelnia levels cannot be counted reliably by Ini
`croscopy below 50;’p.l [corresponding to -
`l[lll,00l).l)00 parasites in the body olan
`adult}. The M'PC is the lowest blood, plasma, or free plasma cm Icerltrnlion which
`produces Ille l11:L\‘ll11tIl1‘| pautslllcitlal eilecl {i.e.. Ill-Ell‘laJ{llI11ll11 parasite reduction
`n1n'ol.‘lbis corresponds to the conccntratlort associated willi firs] slowing ollhe
`lirst order (log linear) decline in pamsiteinia.
`
`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 pharmacodynarriics and how Pl'C—Pl) relation-
`ships should be assessed.
`
`PHARMACOKINETICS
`
`The pharrnacoltinetic (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 lipopltilic. are poorly absorbed after oral or in—
`tram uscular administration and show wide interindividnal differ-
`
`ences in coiicentration profiles. In general. this variation in blood
`concentrations is inversely proportional to bioavailability, which
`emphasizes the importance of improving bioavail-ability 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 closing 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 ‘I2
`
`aac.asn'-torg 5?‘93
`
`lnnoPharma Exhibit 10300002
`
`

`

`Mirlirevlew
`
`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 ofpharrnacokinetic 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—b:-ised 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, cliildren, 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 developcnent. 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 Pl{—PlJ information is enough to decide upon a dosage
`recommendation. 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 oflhedrugs. The antimalarial drugs differ in their stage
`specificities of action against malaria parasites. The 8—an1ino—
`quinolines are unusual in killing pre—ei'ythrocy'tic—stage parasites,
`liypnozoites. and mature ganietocytes ofP.falciparrmr but having
`weak activity against its asexual stages ('29). They are more active
`against asexual stages of P. vivnx and P. lcmiwlesi. All other anti-
`malarial drugs in current use kill the asexual and sexual stages of
`sensitive P. vii-tract, P. mrtlnrirtc, P. ovals, and P. lcriowlcsi and the
`asexual stages and early gatnetocytes (stages 1 to lll_'J ofsensitive P.
`}'ulci'pttr‘tt.-n. but they do not kill the mature P. jitir.‘iptti'rrrn gameto-
`cytes (stage V). The artemisinins have a broader range ofeffect on
`developing P. ,-‘Erlcfpnrmrr sexual stages, as they also kill stage IV
`and younger stage Vgametocytes. Atovaquone and the antifols kill
`preerythrocytic stages and have spororitocidal 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. (mile hypnozoites.
`Even within the asexual cycle there are differences in antimalarial
`activity in relation to parasite developltierit. 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 (30). 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-
`rnalarials. notably. some antibiotics with antirnalarial activity.
`
`have greater effects in the second than in the first drugexposed
`asexual cycle (23). The pharmacokinetiopharmacodynamic rela-
`tionships {PI<—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, 31). Rapid killing ofyoung
`P. jnlcipumm 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—forrn killing is also important, as it contributes to
`the speed ofpatient recovery, but the main tlierapeutic objective is
`to reduce para site multiplication. Once zuitimala rial treatment is
`started. then. after a va_1'i:-:ble lag phase, parasite killing in vivo
`approximates to a first-order process (32-34) as represented by
`the following equation:
`
`P, — n,.~.‘ tr
`
`(1 i
`
`where P, is the parasitemia level at any time I after starting treat-
`ment, PL, is the parasitemia level immediately before starting treat-
`ment. and kg, is the first—order parasite elimination rate constant.
`The parasite clearance halt? life is therefore 0.693119. In equation I.
`parasite killing equates with parasite removalfrom the circulation.
`but in falciparum malaria (but not the other malariasl 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 ancl not drug effects (33). Parasite killing can be ex-
`pressed as the parasite reduction ratio {P1111}. which is the frac-
`tional reduction in parasite numbers per asexual cycle. or the
`reciprocal of ring-form kg. per cycle (32). This cancels out the
`effects ofcytoadherence, as the parasite populations are assessed at
`the same stages ofdevelopment separated by one cycle. The shape
`ofthe concentratiomeffect relationship in viva is assumed always
`to be sigmoid. as it is in vitro (Fig. 2), per the following equation:
`
`ll: _ kin-1.x' lcn/ECSHH l
`
`(-ml
`
`where it is the parasite killing rate and low is the maximum par-
`asite killing rate [i.e., the maximum effect, orfinml for that drug in
`that infection. C is the concentration of drug in blood or plasma.
`ECEU is the blood or plasma C0]1C€11lI_'l'ElT_'lDl1 resulting in 50% of the
`maximum effect, and r1 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 subinaxinial effects in some pa~
`tients} (12). So while concentrations exceed the minimum para-
`siticidal concentration l_MPC).lc in equation I is equal to knm. 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 pl1ar—
`macokinetic—pharrnacodynamic relationships" section below).
`For drugs in current use, maximum PRRs range from approxi-
`
`5?94 at-1Last1'I.arg
`
`Antlmicrobial Agents and Chemotherapy
`
`
`
`isanfisq,r_|,oz‘gtMenueruo,lfiJO'LUSE‘OEE;‘_.tId1J,l.|won|‘.'J9pE0|UMOG
`
`lnnoPharma Exhibit 10300003
`
`

`

`
`
`Leg concentration
`
`FIG 2 Tilt’ co|1cent'rat'ion ellect relationship; for antintztlarial drugs, the effect
`is parasite killing, which can be Ineastlred in tlifferettt ways. The F.I1Ifl.X is the
`maxirnttni parasite killing lliat :1 drug c:1n produce. wliiclt tmrislutes in vivo into
`the inaxiintmt parasite redui.'tinn ratio. The EC5", is the blood or plztsnta con
`cent ration providing 50% ot'n12n:i1n1nn killing. The median and range values
`for a ltyptithelical ptipltlat ion nfnialaria parasites are sltmvn in blue. and Ill:
`clislribtlliolt ufztverztge drug levels in patients is shown as a rcxl be|l—sl|apt-cl
`curve l i.e., cnnct-nt'raI:ions are log normally distributed}. Clearly, sonic of lhe
`patients ltave average drug levels below the MPG and would not have maxi
`nitltti responses with this close regimen.
`
`rnately 10-fold to approximately l0,0t.l0—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 rnonotherapies 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—bion1ass 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 virro method
`assesses a slightly different section ofthe asexual life cycle, which
`may result in important differences between methods in the re-
`sults 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 I 1-8,
`19, 35). Neither is the relationship between inhibition of parasite
`growth and subsequent inhibition of multiplication well estab
`lished. inhibition of growth is measured in most in vitro 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 concentratiomeffect relationship. for predictive mod-
`eling purposes the slopes ofthe linear segments of the in vitro and
`in viva 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-
`sessing blood~stage effects is the lun|.iound,l fraction in plasma.
`Total red cell concentrations are less informative as the parasitized
`
`Minireview
`
`red cells behave very differently from their unparasitized counter-
`parts. ln 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 {S6}. Human malaria infections in irnmuno—
`deficient mice allow PK—PD characterization and thus provide
`useful information in predicting therape utic 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-in susceptibility measures
`l_'S0°/ii inhibitory concentrations [[C50I,
`lC._,.,_,, etc.) and in viva
`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 hi
`the United States during and shortly after the Second World War
`(Fig. 3]. Studies were conducted to determine tl1e optimum dos-
`ing strategies for mepacrine (atebrine, quinacrine), the Cinchona
`alkaloids, and both the 4- and S—arni:noquinolines (3-7-39). Phar-
`macokinetic analysis had yet to he invented, and methods for
`quantitation ofdrugs 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. frtlrriprtritrrt or P. vivax received dilterent 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 antinialarial effects of the four
`main Cinchona alkaloids (quinine, quinidine, cinclionine, 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 ofatltimalarial t1‘eatment [8, 9, 4043).
`(i) PK-PD correlates. Studies ofPK—PD relationships for anti-
`bacterial effects have shown that for some antibiotics (those with
`steep concentration-et'fect 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 ofthe
`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 li.e., the
`higher the Cm“, the larger the All C and the longer the time above
`the MIC]. With some adjustments. these PK measures can be ap-
`plied to antimalarial effects {S2}, although correlates with parasite
`
`
`
`isanfiitq,r_|,oz‘gtiuenueruo,lfiJO'tUSE'OEEflZd1J,l.|moi;pepeoiumoq
`
`
`
`
`
`December 2013 Volume 5}‘ Number ‘I2
`
`aac.asn'-morg 5795
`
`InnoPharma Exhibit 10300004
`
`

`

`Minireview
`
`Plasma
`
`quininemg/L
`
`Effect
`
`0.1
`
`0.2
`
`0.3
`
`0.4
`
`0.5
`
`Daily dose {Grams}
`
`Plasma concentration {r'rIg!l.}
`
`‘l0
`
`WC 3 Dose—responsc relationsliips oblziinecl between lhe years [9115 and 19-16 for quinine in l3lDU(l-llIICll1L'|?(l vivrtx malaria il\‘icCoy strain} in volunteers (.53)-
`Plasrna cmlcenlrulinlis after protein precipiialirnl were menmrcd spectropliotonielricnlly, which overestirmiles parenl compouliti concenlralions. The left but
`shows the variable rt*la1'ionsl1ips he-tween dose and nicali plasma t'oI1cer1tral'ions. and the right graph shows the coliccritraljtiri effect‘ relal'iunsl1ip divided into
`three eff;-cl Iiieasurcs: class 1. no certain ellcct; class 1], lcinporary siipprcssioli of parasitt-min anclior fever: class III, “perInanem" effect.
`i.-2.. absence ol
`parasilenlia for M days.
`
`killing have not been established for most antirnalarial drugs.
`Whereas most bacteria replicate every 20 to 40 min, asexual rna~
`laria parasites infecting humans replicate every I to 3 days. Syrup»
`tornatic infections usually comprise one predominant brood of
`malaria parasites, but multiple genotypes are often present—par—
`ticularly in higher—transmi5sion settiI1gs—and so within one host
`there may be subpopulations with different drug susceptibilities
`(and alsfl diflerent stages of asexual development). The lowest
`blood. plasma, or free plasma concentration which produces the
`maxim Lll'I'l 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 indi\«"idu.=.tl’s
`
`infection. Innate host—det'ense mechanisms and acquired irnni une
`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 110l1l.IIlIIllJl}€ patients 44). has not been well char-
`acterized.
`
`with current dosing for all antimalarial drugs except the arte~
`misinin derivatives. drug elimination is sufficiently slow that the
`antirnalarial efiects ofa treatment persist for longer than one asex—
`
`Concentration
`
`Weeks
`
`FIG 4 Plasma or blood concuntra Lion profile of a slowly eliniinalud a.nti.ma
`larial drug showingnn arbitrary MIC. The AUC is the area under the c11rve,ancl
`Cnlax is Ihe lI'lfl..\'ilIllIll'l colicenlration in blood or plasma. AUC from 7 days to
`infinity is shown in darker pink. Blond conccnI1"at_ions are increasingly mea-
`sured on day Sr’ in thcrapcutiu.‘ assessments of slowly eliminated antirnalarials
`H9).
`
`Total
`
`parasites
`
`Weeks
`
`FIG 5 l'Jif'i'ert-I11 tlieraipeulin: responses to .1 slowly eliminated anliilialarial
`drug in at rIla.l:iri:J infeclirnl of I01" p:1rasiles {parasite density,
`-2,(l(l(l,i|.Ll). The
`blood concciitration profile in gray is shown in the backgrouml. Parasitologi
`cal responses range from fully sensiriw {green} to highly re.sisia1il.(blw.‘}. Each
`respunse is associated wil h :1 dillerelil level of snsce}: Iibili ty and lhus a diilerenl
`MIC and MPC [arrows polrnlillg, to collceniraliorn profile}. The inset repre-
`sents the coriceiitratitsii ell}-cl. relatioiishjp tor lhc lowesl. level of resistance
`(resulting in it late failure). showing -:oi'respni'ICling points for the 1\‘flC and
`h-'iPC(or;1nge curve}.
`
`
`
`
`
`isanfiliq,i_|,oz‘gtMenueruojl3JO'LUSE'OEE[fZd11l.|moi;papeoiumog
`
`5?96 aacasrruorg
`
`Antimicrobial Agents and Chemotherapy
`
`lnnoPharma Exhibit ‘l030.0005
`
`

`

`ual cycle (3, 22, 32. 45). Indeed, many antimalarials have terminal
`elimination half—lives (r, 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 ifdrug 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 conceiitratiomeffect 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 o

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