throbber
232
`
`Part III I Clinical Testing
`
`5. PHARMACOKINETICS VS PHARMACODYNAMICS
`It is imperative to consider not simply the pharmacokinetics of a new agent, but its
`overall pharmacodynamics. Relatively few clinical studies have examined the relation(cid:173)
`ship between therapeutic, or toxic, response drug dose and exposure time (12).
`Rather, a great deal of emphasis has been placed on defining the interdependent rela(cid:173)
`tionship of concentration and time on antitumor activity (13). Much conflicting data
`addressing the complex relationship of antitumor activity to extracellular drug con(cid:173)
`centrations (q vs exposure time (f) exist. Several investigators have determined that
`the cytotoxicity profile of many agents, such as methotrexate, 5FU, or adriamycin,
`was not a simple function of C x T (14-16), whereas others convincingly show a
`strong dependence of activity on C x T (17-20). Skipper and Adams have suggested
`that new drugs could be developed based on a differential concentration coefficient
`for tumor cell kill vs host toxicity (12,21). This hypothesis is a direct extension of the
`established principle governing bacterial disinfectant action: survival as a function of
`Cn x T (12). Specifically, log transformation of concentration-time curves yields a
`line whose slope is defined as the concentration coefficient, n, and whose intercept is
`the exposure constant, k, for that level of cell kill. Although n may approach unity
`for many agents and thereby demonstrate direct dependence on C x T, many others
`may deviate significantly from unity, providing a very different relationship between
`activity and C x T. This particular pharmacodynamic principle would take into
`account other complex processes that introduce heterogeneity not taken into account
`by the simple C x T model. When this principle was applied to agents that did not
`conform to the simple C x T equation, it was shown that the data conformed to the
`en X T model. Pinedo and Chabner concluded that methotrexate depletion of
`nucleated murine bone marrow cells in vivo was not a simple function of C x T (16).
`By reanalzying their data employing this principle, Adams determined an n of 0.33
`(R 2 = .992) which indeed conformed to the Cn x T parameter (12).
`This pharmacodynamic principle also provides a potential parameter for comparing
`antitumor agents. This parameter represents the minimum exposure conditions re(cid:173)
`quired for a specific level of antitumor activity and accounts for differences in concen(cid:173)
`tration coefficients among agents. Adams has demonstrated this principle elegantly in
`his examination of Crisnatol, a new DNA intercalator in comparison to other agents,
`including doxorubicin, 5-FU cisplatin, etoposide, and tamoxifen. Analysis of these
`agents in the MCF-7 model shows that the minimum en x T parameter gives a relative
`cytotoxicity profile distinct from that found with the standard IC,. end point (12).
`
`6. THE STARTING DOSE
`The initial starting dose is a critical parameter in the design of any Phase I trial.
`Anticancer drugs are inherently toxic agents operating in a very narrow therapeutic
`window. Thus, an aggressive starting dose can result in irreversible toxicity and mor(cid:173)
`tality, leading to a premature tabling of a potentially useful agent. On the other hand,
`a particularly conservative initial dose will lead to poor utilization of resources and ex(cid:173)
`cessive prolongation of a trial. Ideally, a Phase I study should not require more than
`six escalation steps (22). Furthermore, given that patients in the study are treated with
`therapeutic intent, many will be given homeopathic or subtherapeutic doses.
`
`;
`i"!I
`~,
`
`\l
`!
`:~
`11-•
`' _ti"~ Ii
`·~
`I~ • \
`
`Chapter 12 /Phase I Trial Design
`
`233
`
`The starting dose on a Phase I agent is generally extrapolated from preclinical tox(cid:173)
`icology data obtained in rodents, dogs, and less frequently, primates. Typically,
`single- and multiple-dose treatment schedules are employed. Despite the inherent
`problems and assumptions generalizing from animals to humans, a safe and accurate
`starting dose can usually be determined. Interspecies comparisons of dose are most
`• This basic tenet of Phase I trial design
`reliably based on body surface area or mg/m2
`stems from elegant work performed by Pinkel and Freireich, who showed that the
`toxic dose of an agent is similar among species when the dose is measured on the basis
`of body surface area or mg/m2 (23,24). This parameter, unlike body weight, appears
`to have a tighter correlation with physiological functions and inter- and intraspecies
`(adults vs children) toxicity thresholds (24).
`The use of preclinical toxicology studies in multiple species for all anticancer agents
`dates back to the inception of the Drug Development Program at the NCI in the
`mid-1950s. In 1973, the Laboratory of Toxicology, Division of Cancer Treatment,
`NCI, developed guidelines for preclinical toxicology testing of cancer chemothera(cid:173)
`peutic agents (25). The requisite studies were extensive, and involved single-dose
`schedules to assess acute toxicity and lethality in dogs, monkeys, and rodents. Sub(cid:173)
`acute toxicity was determned on dogs using multiple-dose schedules. Following a pre(cid:173)
`determined observation period, the animals underwent a full histological evaluation
`to determine the drug's toxicity profile. The starting doses for a clinical trial of a
`Phase I agent were generally derived by calculating one-third of the lowest toxic dose
`for the most sensitive species (dog or monkey).
`The preclinical toxicology studies were streamlined in the late 1970s, when inter(cid:173)
`species toxicity comparisons could be made because of the extent of toxicology data
`compiled in animals and humans. In October of 1979, the Food and Drug
`Administration Oncologic Drugs Advisory Committee agreed that preclinical studies
`in the mouse could predict quantitative drug toxicity in humans. Thus, the starting
`dose for clinical trials was established as s 1/10 the LD,. (the lethal dose to lOO!o of
`nontumor-bearing animals), determined from statistically reliable murine lethality
`studies. For validation of the human starting dose and to compile further qualitative
`toxicology data, the beagle dog was the species chosen for additional preclinical
`testing (25).
`In 1980, toxicology protocols were developed by the Division of Cancer Treatment
`incorporating these recommendations. Typically, lethality studies are performed on
`mice in single (daily x 1) and multiple dose (daily x 5) schedules, and establish the
`dose causing lethality in 10, 50, and 90% (LD,., LD,., and LD,.) of animals tested
`(25). The LD,. dose is converted to mg/m2 and 1/10 the equivalent dose or 1110
`MELD, 0 is given to the beagle dog. If no toxicity is encountered, the dose is escalated
`until minimal reversible toxicity, i.e., the toxic dose low (TDL) is noted in the dog.
`The TDL has been defined as the lowest dose that produces drug-induced pathologic
`alterations in hematologic, chemical, clinical, or morphologic parameters; doubling
`the TDL produces no lethality (5,26). The human equivalent of 1/3 the TDL in dogs
`in mg/m2 would then be the starting dose in Phase I clinical trials. Conversely, if the
`1110 MELD,. is not tolerated in the dog, the dose is reduced until the dog TDL is
`established. This approach yields a statistically more reliable toxicology profile
`because a larger number of mice in comparison to dogs can be studied.
`Grieshaber and Marsoni subsequently performed a retrospective analysis on seven
`antineoplastic agents that had completed clinical Phase I trials to determine the cor-
`
`
`
`19 of 36
`
`Celltrion, Inc. 1053
`Celltrion v. Genentech
`IPR2017-01122
`
`

`

`!t ;; \(
`'i .t
`
`'.f
`• ,l.
`l
`
`1
`
`' ~
`
`!
`'I
`
`!'
`;

`1t
`1k
`
`I ~ ~·
`
`~
`, I~
`'I·
`'t· ,,.
`
`t
`I ~-
`;'
`
`234
`
`Part III I Clinical Testing
`
`relation of preclinical toxicology findings to the DLT in humans. They showed that
`1/10 MELD,. reliably predicted a safe human starting dose in 6/7 drugs tested,
`whereas the beagle dog toxicology data had little predictive value in establishing the
`starting dose. However, the preclinical toxicology studies in dogs revealed the human
`DLT IOO"lo of the time with 7SOfo accuracy in providing TDLs. The preclinical mouse
`studies revealed that DLT 7SOfo of the time with SOOfo accuracy in predicting TDLs in
`the human (25). Thus, the beagle dog appears to be a much better model for predict(cid:173)
`ing toxic effects in the human, whereas the mouse may be a better model for
`establishing an initial starting dose.
`Similarly, Goldsmith et al. analyzed mouse, dog, and monkey data on 30 agents
`for which there was correlative Phase I qualitative and quantitative human toxicity
`data (27). However, using 1/3 the TDL in dogs would have underpredicted the
`qualitative toxicity of the drug in 6/30 drugs analyzed. Similarly, Homan's analysis
`indicates a S .90fo probability of exceeding the safe human dose if the 1/3 TDL rule is
`applied to every drug (28). Additional valuable information was drawn from rodent
`toxicity data that would have underpredicted the toxicity of the agent in only 2/29
`cases analyzed. In their retrospective analysis of 12 Phase I agents, Penta et al. have
`also confirmed the potential utility of murine toxicology data predicting an accurate
`and safe starting dose for Phase I chemotherapy trials (29). These data suggest that
`the starting doses in clinical studies should not be based exclusively on the 1/3 TDL in
`the most sensitive large animal and speak to the importance of collecting comparative
`interspecies pharmacokinetic data. There can be significant interspecies differences in
`absorption, distribution metabolism, and excretion of drugs resulting in both quan(cid:173)
`titative and qualitative differences in pharmacodynamics (27). The analysis of such
`comparative data would be significant in determining the human starting dose.
`The rat is also being used in preclinical toxicology studies, since laboratory data
`can be collected with greater ease in comparison to its smaller murine cousin. In fact,
`the European Organization for Research and Treatment of Cancer (EORTC) and the
`United Kingdom Cancer Research Campaign Phase I/II Trials Committee (UKCRC)
`has designated the rat rather than the dog as the second species to confirm the murine
`LD,. (30).
`Despite the utility of preclinical toxicology data in establishing qualitative effects of
`new agents, there are limitations. Although preclinical studies have good predictive
`value for bone marrow, gastrointestinal, hepatic, and renal toxicity, they predict
`poorly for dermatological and cardiac side effects. Moreover, owing to the difficulty
`in performing adequate neurological evaluations in animals, preclinical studies pre(cid:173)
`dict unreliably for central and peripheral nervous system toxicity (23,31).
`
`7. DOSE ESCALATION
`The issue of dose escalation in a Phase I trial is complex, stemming from the
`relative lack of knowledge regarding dose and probability of toxicity for novel anti(cid:173)
`cancer agents. Typically, anticancer agents operate in a very narrow therapeutic win(cid:173)
`dow following nonlinear kinetics, and thus, the potential for life-threatening toxicity
`at effective dosage levels is significant. For this reason, ethics would dictate that the
`agent cannot be tested in a normal healthy volunteer, but must be examined in a
`cancer patient with refractory disseminated disease for whom no other therapeutic
`
`Chapter 12 I Phase I Trial Design
`
`235
`
`Table 3
`Modified Fibonacci Dose
`Escalation Scheme for Phase I Trials
`Percentage increase
`above preceding
`dose level
`
`Drug dose
`n
`2.0n
`S.On
`7.0n
`9.0n
`12.0n
`16.0 n
`
`100
`67
`so
`40
`30-35
`30-35
`
`option exists. The examination of Phase I agents in this study population pose many
`ethical dilemmas that impact directly on the choice of a dose escalation scheme. Since
`therapeutic intent underscores all Phase I trials, it would follow that using a conserva(cid:173)
`tive schema will result in patients receiving subtherapeutic or ineffective doses. Con(cid:173)
`versely, employing an aggressive dose escalation model will lead to the greater
`likelihood that patients will experience toxicity at or beyond the allowable level.
`
`7.1. The Modified Fibona.cci
`Initially proposed by Schneiderman (32), the modified Fibonacci method is an em(cid:173)
`pirical approach to dose escalation that has become the standard methodology in
`Phase I trials. It is based on the Fibonacci sequence, which begins with the integers 1,
`1, 2, 3 and so on, with each number representing the sum of its two predecessors. Let
`n mg/m2 be the dose received by the initial group of patients. In the Fibonacci scheme
`of dose escalation, successive groups of patients receive 2n, 3.3n, Sn, 1n, 9n, 12n,
`16n, and then doses increase by a third of the previous dose (Table 3) (22). In
`other words, this method uses successive percentage increases above the preceding
`dose level of 100, 67, SO, and 40 with subsequent increments of 30-3SOfo (5,33). The
`modified Fibonacci scheme thus slows the Fibonacci sequence, so that a greater
`percentage of increase in the dose escalation occurs earlier in the trial. Obviously, if
`preclinical toxicity studies demonstrate a steep dose/toxicity curve, dose escalation
`would be more conservative than that dictated by the Fibonacci schema.
`Although not always ideal in efficiently predicting an accurate MTD, i.e., the true
`dose that would yield a specified levd of dose-limiting toxicity, the modified Fibonacci
`generally yields a fair approximation of the MTD while operating in a range of safety
`for the majority of patients enrolled (2,22,27,34). Goldsmith et al. have examined the
`efficiency of the modified Fibonacci in the context of the number of dose escalations
`required for 28 different Phase I studies to reach the MTD on a particular schedule.
`Using 1/3 the TDL in dogs as the human starting dose (after excluding those for
`which the starting dose was too high (vide supra), the number of dose escalations for
`20/28 schedules tested ranged from 1-S, whereas only 8 schedules required more than
`6 dose escalations (range 7-13) (27).
`The typical Phase I design dictates the enrollment of cohorts of 3-6 patients/ dose
`level until a predetermined level (generally 330fo) (35) of toxicity is experienced. This
`
`
`
`20 of 36
`
`Celltrion, Inc. 1053
`Celltrion v. Genentech
`IPR2017-01122
`
`

`

`236
`
`Part III I Clinical Testing
`
`stopping dose level, or more frequently the previous dose level, will then be the MTD
`and the starting dose for most Phase II trials of the drug.
`lntrapatient dose escalation (dose escalation within individual patients) at nontoxic
`dose levels is generally not performed. The rationale for this conservative approach is
`the potential for compromising adequate interpretation of toxicity data. Specifically,
`one could not reliably attribute an observed toxic effect to either cumulative toxicity
`or immediate toxicity from the escalated dose (22,36).
`Underlying the modified Fibonacci approach of dose escalation is a layer of con(cid:173)
`servatism that may result in an inefficient use of patients and resources. This, in tum,
`has significant impact on ethical and practical issues in Phase I trial design and drug
`development. Again, inherent in a Phase I trial design is therapeutic intent, which dic(cid:173)
`tates that patients receive therapeutic or near-therapeutic doses of the Phase I agent.
`Still, this goal must be balanced by the threat of unanticipated and unacceptable tox(cid:173)
`icity from aggressive dose escalation. There have, therefore, been several other pro(cid:173)
`posed methodologies to address the aforementioned limitations of the modified
`Fibonacci strategy.
`
`7.2. Pharmacologically Guided Dose Escalation
`The pharmacologically (or phannacokinetically) guided dose escalation (PODE)
`schema was originally proposed by Collins and coworkers in the Blood Level Work(cid:173)
`ing Groups of the Division of Cancer Treatment at the NCI (37). The foundation of
`PODE rests on the basic concept of toxicity and efficacy of a drug being derived from
`its concentration vs time curve. This concept originated from the work of German
`pharmacologists during World War I (13,21). They observed that mustard agents
`were equally toxic whether inhaled for a short time at a high concentration or for a
`long time at a lower concentration. A narrower application of this principle to anti(cid:173)
`cancer agents with regard to mechanism of action and schedule was performed by
`Skipper et al. Their work demonstrated that the effect of antimetabolites on macro(cid:173)
`molecular systems is more dependent on time of exposure above a certain threshold
`concentration than absolute concentration or C x T (38). ('11te importance of con(cid:173)
`sistency of schedule across species on reliable interpretation of PK data and applica(cid:173)
`bility to the design of Phase I trials was stressed. It is interesting to note that in his
`retrospective review of preclinical toxicity data on 30 drugs [vide supra] in com(cid:173)
`parison with the actual clinical doses and schedules used in humans, only four drugs
`had identical schedules in humans, monkey, dog, and mouse (27). With these prin(cid:173)
`ciples as a basis, Collins proposed that C x T (the area under the curve for plasma
`concentration vs time) would be a more realistic index of toxicity than dose, citing
`three potential explanations for variation in toxicity between mouse and humans
`using dose-based comparisons:
`
`j:;
`
`t-f;i~
`I ·~
`·1:
`I ~ ~~
`~~:·
`i'l
`:Ii~
`~
`·~ I.,
`\~;
`
`l ;~ ti
`l
`.i
`I~
`b!;
`.c./ u; ,i:
`l.
`~ l~i
`. 1.1
`,}p.
`1;~
`·~~
`j:"
`1.lf
`Is:
`Le
`i';.
`!
`l•I
`I·~
`'l \
`
`'4
`'11
`
`1
`
`I. Species differences in drug metabolism, elimination, and binding;
`2. Schedule dependency owing to exposure time differences; and
`3. Species differences in target cell sensitivity.
`
`'•
`
`Collins constructed a pharmacodynamic hypothesis that similar biological effects
`(e.g., toxicity) would occur at similar plasma levels in mice and humans (39). In other
`words, the ratios of C x Tin human plasma at the MTD to C x Tat the LD, 0 of an
`
`Chapter 12 I Phase I Trial Design
`
`237
`
`agent should approximate unity if this hypothesis is valid. Retrospectively, Collins
`analyzed 13 anticancer agents for which there were comparative pharmacokinetic and
`pharmacodynamic data. In 10/13 drugs examined, the hypothesis was correct with
`C x T ratios ranging from 0.6-1.3, demonstrating near-equivalent toxicity. The
`other three agents did not conform to the model presumably because of interspecies
`differences in target organ sensitivities (39) (vide infra).
`In addition to using interspecies PK data for efficient dose escalation, Collins
`stressed the importance of examining preclinical quantitative data on schedule
`dependency with regard to its impact on accelerated entry dose in Phase I trials. In
`addition to total drug exposure, the rate of drug delivery can have a significant effect
`on the toxicity profile of a drug. Collins attributes this relationship to a "threshold"
`or peak-level phenomenon (39). The toxicity incurred by a rapid delivery or bolus ad(cid:173)
`ministration of a drug may be abrogated by a longer or continuous infusion. Such a
`schedule would permit greater drug delivery without the toxicity from high peak
`concentrations. The PODE approach can be optimally applied to drugs given on a
`continuous infusion schedule by replacing C x T values or AUC by measured steady(cid:173)
`state concentration of the drug. Steady-state concentration targets are more easily
`grasped and avoid the difficulties of exposure-time differences caused by pharmaco(cid:173)
`kinetic factors (13,40).
`Collins prospectively applied the pharmacodynamic and pharmacokinetic prin(cid:173)
`ciples to eight agents consecutively entered into Phase I trials under NCI sponsor(cid:173)
`ships. Three of the eight agents were too far along in trial to benefit from this pharma(cid:173)
`cologically guided method. Nevertheless, the remaining five trials were significantly
`shortened either because of an increased entry dose or accelerated escalation schema
`(39). For example, merbarone demonstrated an LD,. in mice based on bolus adminis(cid:173)
`• This translated into a starting dose of 12 mg/m2. However,
`tration of 123 mg/m2
`because of acute toxicity noted, continuous infusion schedules in dogs were explored,
`resulting in a revised starting dose of 96 mg/m2/d. Since the human MTD was 1500
`mg/m2/d, the eightfold-higher entry dose resulted in 48 fewer total patients being
`accrued and the study being completed 16 mo earlier than initially anticipated.
`Similarly, for flavone acetic acid, the entry dose was based on the standard 1/10 the
`LD,., but dose escalation proceeded more rapidly based on plasma levels, thereby
`saving four unnecessary Fibonacci steps (39). Assuming the C x T ratio was an effec(cid:173)
`tive predictor for toxicity, Collins concluded that the PODE method could result in a
`20-500/o reduction of dose escalation steps.
`Intrigued by Collins' observations and proposals, the Phannacokinetics and Metab(cid:173)
`olism (PAM) group of the EORTC (now the EORTC Pharmacology and Molecular
`Mechanisms Group) further evaluated the PODE method by retrospectively cor(cid:173)
`relating their extensive human and murine phannacokinetic data with known toxicity
`data in the Phase I setting (37,41). A careful review of their data identified problems
`with this methodology, but provided encouragement for the further evaluation of
`PODE in Phase I study designs. Cited technical problems included PK data obtained
`in animals using disparate routes of administration compared with the human data,
`the lack of an accurate LD,. in mice, and the use of doses other than the LD,. for
`pharmacokinetic studies. Cited pharmacokinetic factors that would affect the basic
`tenet of PODE approach, i.e., toxicity being solely a function of total parent drug
`plasma levels, included:
`
`
`
`21 of 36
`
`Celltrion, Inc. 1053
`Celltrion v. Genentech
`IPR2017-01122
`
`

`

`Chapter 12 /Phase I Trial Design
`
`239
`
`data alone. Instead, its value should be considered in the wider context of deploying
`pharmacokinetic and pharmacodynamic information to optimize and shape early
`clinical trials.
`
`7.3. Other Statistical Strategies
`Additional methodologies to address the shortcomings of the standard Phase I
`design included several statistical approaches, including variations on escalation/
`de-escalation schemes ("up and down") (42-45), stochastic approximation methods
`(44,46), and the application of Bayesian principles to estimate more accurately the
`probability of toxicity at the Mm (35,47,48). Bayesian methods are based on assump(cid:173)
`tions about the main end point of the study, known as "priors," which are derived
`from the investigators' prior observations and/or beliefs stemming from their own
`experience and that of others (49). An example of a classic Bayesian approach that
`proposes model-guided dosing includes the Continual Reassessment Method (35,
`48,50). Using such an approach, information from preclinical studies and/or clinical
`studies of similar drugs is used to make an educated guess regarding the dose-toxicity
`curve and the recommended Phase II dose. Patients are treated at the current esti(cid:173)
`mated recommended Phase II dose that is continually updated to reflect the accumu(cid:173)
`lating dose-toxicity data. When the predetermined sample size has been reached, the
`final estimate of the recommended phase II dose is made from all available data (49).
`A potential drawback of this approach is overestimating the starting Phase I dose,
`thereby placing the initial patients enrolled at great risk for drug toxicity. The Quanti(cid:173)
`tative Assessment Phase I Escalation Study Design, a related approach, uses an itera(cid:173)
`tively updated pharmacodynamic model for dose escalation. Unlike the Continual
`Reassessment Method, dosing is more dependent on true observations of toxicity
`rather than assumptions or "priors." The scheme utilii.es a cohort-based escalation
`approach similar to that used in a traditional Phase I design. As dose-toxicity data
`are accumulated, a pharmacodynamic model is fit to the data. Model-guided dose
`escalation begins only after a specific number of cohorts of patients have been observed
`for toxicity and a dose-toxicity relationship has been statistically defined (49,51). This
`strategy, although currently applicable only to drugs with myelosuppression as the
`DL T, shows significant promise in more accurately and efficiently defining the Mm
`in Phase I studies of myelosuppressive agents. With modification for nonhematologic
`toxicity, such a scheme could have more widespread application.
`
`7.4. Patient Choice Dose Cohort
`A more innovative approach to Phase I design incorporating issues influencing
`patient participation has been proposed by Daugherty et al. This design incorporates
`such less-well-defined aspects as patient motivation and comprehension and the influ(cid:173)
`ence of patients' aversion to risk. In this particular scheme, the patient would play an
`integral role in determining his or her entry dose level.
`Using the Patient Choice Dose Cohort (PCDC) Phase I trial design, patients choose
`from one of three possible doses:
`
`I. The standard Phase I trial starting dose of 1/10 LD .. in mice;
`2. Some multiple (M) of 1110 LD .. ; or
`3. Twice the (M) dose.
`
`i
`~
` f
`t'
`)
`\;.
`
`Ii
`
`l
`~
`j
`\.
`
`~ I '
`
`n
`
`If
`
`~ i
`~
`~
`ii Ii
`M u
`ii 1 I~
`11 Ii
`I;
`
`II
`
`•i
`
`238
`
`Part III I Clinical Testing
`
`1.
`
`Interspecies differences in plasma protein binding resulting in disparate levels of free
`drug and hence different toxicity profiles;
`2. Differences in metabolism of the drug that may affect levels of active drug in the case of
`a prodrug that requires metabolism to the active form or may impact on levels of toxic
`metabolites;
`3. Nonlinear pharmacokinetics in which a small increase in dose would have a significant
`effect on AUC;
`4. Patient heterogeneity resulting in significant variance of measured AUC; and
`S. Lack of adequate PK data on tissue drug levels that may more accurately reflect the
`pharmacodynamic effect of a drug.
`
`Interspecies differences in target organ or cell sensitivity were also stressed relative
`to observed disparate pharmacodynamic effects. Collins cited antimetabolites as an
`example of a class of agents that are not amenable to the PGDE approach because of
`the differences in target cell sensitivity. Antimetabolites are highly dependent on the
`target cell for activating or inactivating enzymes and extracellular nucleic acid precur(cid:173)
`sors. As such, antimetabolite toxicity cannot reliably be predicted by AUC or dose.
`Despite the shortcomings of the PGDE method, the EORTC concluded that it was
`valuable especially when applied to certain classes of drugs. In particular, the anthra(cid:173)
`cyclines demonstrated AUC to be a better predictor of toxicity than dose. Similarly,
`platinum agents showed a reasonable correlation between AUC and toxicity. As
`anticipated, several N-methyl drugs that require metabolism to the active species
`demonstrated poor correlation between AUC and toxicity.
`Graham and Workman critiqued the PGDE approach applied prospectively to
`several new anticancer agents, including amphethinile, brequinar sodium, iodo-doxo(cid:173)
`rubicin, the anthrapyrazoles, rhizoxin, and aphidicolin glycinate (37). Although their
`review will not be detailed here, their analysis supported the global value of this ap(cid:173)
`proach in providing critical information for the rational design of Phase I trials. As
`demonstrated, the PGDE was not rigidly applicable to many agents, but did provide
`information that often gave valuable insight into a new drug's pharmacologic effect
`and clinical potential. For example, in a prospective Phase I study of iodo-doxorubi(cid:173)
`cin, an anthracycline derivative, a PGDE strategy was attempted, incorporating
`Collins' proposals and the guidelines drafted by the EORTC. Near the outset of the
`study, it became clear that marked species differences exist in the pharmacokinetics
`and metabolism of the drug. Principally this was attributed to the rapid metabolism
`of 1-Dox to 1-Doxol by an aldo-keto reductase absent in mice. Thus, although the
`correlative PK studies were modified to measure the metabolite as well, a conven(cid:173)
`tional modified Fibonacci scheme was used. Additional elegant PK investigations
`demonstrated that the toxicity of the parent drug and metabolite had comparable
`LD,. values in mice. As a result of these findings, a PGDE scheme was reintroduced
`and subsequent doses escalated by the summation of the 1-Dox and 1-Doxol AUCs,
`utilizing the original target 1-Dox AUC to guide the dose increments. In addition to
`providing valuable pharmacological data on 1-Dox, by using the PGDE strategy with
`pre-existing knowledge of the comparative pharmacological information on 1-Dox
`and 1-Doxol at the outset of the study, the Mm could have been reached in only five
`steps (37).
`Graham and Workman concluded that the value of this approach should not be
`judged on its ability to predict accurately the MTD based on correlative animal AUC
`
`
`
`22 of 36
`
`Celltrion, Inc. 1053
`Celltrion v. Genentech
`IPR2017-01122
`
`

`

`240
`
`Term
`Subtoxic dose
`
`Minimal toxic dose
`
`Recommended dose for therapeutic
`(Phase II trial)
`MTD
`
`Part III I Clinical Testing
`
`Table 4
`Definltloo of Dose Toxicity lo Phase I Studies
`Definition
`A dose that causes consistent changes of
`hematologic or biochemical parameters
`and might thus herald toxicity at the
`next higher dose level or with prolonged
`drug administration
`The smallest dose at which one or more
`of three patients show consistent, readily
`reversible drug toxicity
`The dose that causes moderate, reversible
`toxicity in most patients
`The highest safely tolerable dose
`
`Once a PCDC had been filled and the patient treated without toxicity, that selected
`PCDC would become the lowest PCDC (X). Subsequent patients would then choose
`from X, MX or 2MX for their dose selection. Of note, Daugherty et al. also proposed
`that cohorts of patients be limited to one, unless a grade III or IV toxicity was
`experienced (52). This method has the advantage of more efficiently utilizing patient
`resources, but a meaningful interpretation of the confidence interval for a potential
`toxicity would be sacrificed.
`In summary, it is evident that additional methodologies to address the inadequa(cid:173)
`cies and shortcomings of the current Phase I trials design must be explored. The two
`factors most frequently modified in the new designs are improving patient safety or
`maximizing the potential for response. However, the very nature of cytotoxics demon(cid:173)
`strating a narrow therapeutic range requires that a change in one factor be accom(cid:173)
`panied by a reciprocal change of the other. Hence, patients who have the greatest
`likelihood of responding to a new anticance agent are also the patients with the great(cid:173)
`est risk for toxicity. As Hawkins has so succinctly stated in his critique of the newer
`methods, "the feasibility of any new trial design must ultimately be demonstrated by
`the successful conduct of the clinical study" (53). Inherent in the definition of success
`is maintaining an ethically reasonable safety zone for patients enrolled in the study.
`
`8. TERMINATING THE PHASE I CLINICAL TRIAL
`
`8.1. De(i.ning The MID
`The definition of the MTD is somewhat arbitrary and varies from investigator to
`investigator. Some define it to be the maximal dose administered during the trial with(cid:173)
`out eliciting any toxicity. Others define the MTD as the minimal administered dose
`associated with a toxic response. These definitions, however, arc strongly dependent
`on sample size. Storer takes the MTD to mean some percentile of the dose toxicity
`relationship, about the 33rd (43). More generally, estimating some arbitrary percen(cid:173)
`tile chosen by the experimenter seems desirable (44).
`
`8.2. Toxicity Grading System
`Reasonable definitions of toxic levels in humans have been established by Carter
`et al. and shown in Table 4 (2,33). However, the TDL and the MTD in human trials
`
`,,~
`
`4
`ff
`p
`I
`~
`~ ~
`u ~ 4
`
`IT
`~
`
`~ e
`
`I &
`'
`~ w i
`9 " ~
`
`I ' i
`
`~ j
`)i
`~ u

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