throbber
E U R O P E A N J O U R N A L O F C A N C E R 4 5 ( 2 0 0 9 ) 2 7 5 – 2 8 0
`
`a v a i l a b l e a t w w w . s c i e n c e d i r e c t . c o m
`
`j o u r n a l h o m e p a g e : w w w . e j c o n l i n e . c o m
`
`Optimising the design of phase II oncology trials:
`The importance of randomisation
`
`Mark J. Rataina,*, Daniel J. Sargentb
`aSection of Hematology/Oncology, Department of Medicine, Committee on Clinical Pharmacology and Pharmacogenomics,
`The University of Chicago, Chicago, IL, United States
`bDivision of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, United States
`
`A R T I C L E I N F O
`
`A B S T R A C T
`
`Article history:
`Received 17 October 2008
`Accepted 29 October 2008
`Available online 6 December 2008
`
`Keywords:
`Phase II
`Clinical trial design
`Randomization
`
`Oncology trial end-points continue to receive considerable attention, as illustrated by the
`development and revisions to the RECIST criteria. In this article, we focus the reader away
`from the issue of end-points for phase II trials and towards what we believe to be an even
`more important issue, the fundamental need for randomisation in phase II oncology trials,
`ideally with blinding and dose-ranging. We present arguments to support the proposition
`that randomisation will enable greater clarity in the interpretation of the phase II trial
`results, as well as allowing for more precise estimates of the effect size and sample size
`requirements for definitive phase III trials. Randomisation will also reduce potential bias
`resulting from inter-trial variability, which inflates both type I and II errors if historical con-
`trols are utilised. In the context of a randomised blinded trial, the exact choice of end-point
`is less critical, although we favour end-points such as the change in tumour size or progres-
`sion status at a fixed early time point (i.e. 8–12 weeks after randomisation). Although end-
`points based on RECIST criteria can and should be utilised in randomised phase II trials, we
`do not believe that revision of the RECIST criteria will result in a fundamental improvement
`in drug development decisions in the absence of randomised clinical trials at the phase II
`stage of drug development.
`
`Ó 2008 Elsevier Ltd. All rights reserved.
`
`1.
`
`Introduction
`
`The phase II clinical trial plays a central role in oncology drug
`development. After a phase I trial has determined a tolerable
`dose for a new agent or combination, a well-designed phase II
`trial should provide the information required to make a go/
`no-go decision regarding subsequent phase III testing. As
`phase III trials require several years, hundreds or thousands
`of patients and often tens or hundreds of millions of dollars,
`the information that a quality phase II trial can provide is
`essential to a decision regarding the potential investment in
`a larger trial. In this paper, we present a rationale for the ex-
`
`panded use of randomisation in phase II oncology trials, in or-
`der to better inform this decision-making process.
`
`2.
`
`Why do we do phase II trials?
`
`Phase II trials should be most appropriately viewed as proof of
`concept trials, used for the purpose of determining whether a
`particular agent (or combination) should be studied further.1
`In this sense, they serve a critical filtering mechanism, in
`which a negative trial should lead to the discontinuation of
`development of a new agent for the selected indication.
`Optimising the filtering process is the critical issue: too tight
`
`* Corresponding author: Address: 5841 S, Maryland Ave., MC 2115, Chicago, IL 60637, United States. Tel.: +1 (773) 702 4400; fax: +1 (773) 702
`3969.
`E-mail address: mratain@medicine.bsd.uchicago.edu (M.J. Ratain).
`0959-8049/$ - see front matter Ó 2008 Elsevier Ltd. All rights reserved.
`doi:10.1016/j.ejca.2008.10.029
`
`MYLAN - EXHIBIT 1029
`
`

`
`276
`
`E U R O P E A N J O U R N A L O F C A N C E R 4 5 ( 2 0 0 9 ) 2 7 5 – 2 8 0
`
`a filter will terminate promising agents improperly, but a too
`porous filter will result in an excessive number of costly neg-
`ative phase III trials. In the previous era of oncology drug
`development, there were very few drugs available for the
`study, and as such a porous filter was very appropriate to
`minimise the possibility that a false-negative result would re-
`sult in the discarding of a promising agent.
`In the current era, there are hundreds of investigational
`oncology drugs available for the study. As such, we believe
`that it is more appropriate (at least at a societal level) to use
`a tighter filter, one that only advances to phase III develop-
`ment of those drugs for which there is a high probability of
`success in the phase III trial. We recognise that this strategy
`may be problematic for companies with a single drug in
`development. However, from a societal view, minimising the
`number of phase III trials of ineffective agents is important,
`as patients enrolled on a negative phase III trial may have lost
`the opportunity to participate in the trials of active investiga-
`tional agents, and financial and intellectual resources spent
`on a negative phase III trial similarly would be better spent
`on the development of new agents. Given the patient and
`financial resources required for a phase III trial in the current
`environment, we feel that it is more important to minimise
`the risk of failing to demonstrate the efficacy in phase III trial
`than to be concerned about the lost opportunities subsequent
`to not proceeding to phase III testing.
`We propose that attempting to ensure that phase III trials
`attain success on their specified end-point is best determined
`through the explicit modelling of the relationship of dose to
`both efficacy and toxicity. If an identifiable and reliable rela-
`tionship between dose and the chosen efficacy end-point
`can be established, considerable evidence is provided in sup-
`port of the success of the eventual phase III trial, particularly
`
`Table 1 – Impact of randomisation on strength of
`inference in phase II trial.
`
`if that efficacy signal is apparent at doses with an acceptable
`toxicity. On the other hand, if there is no relationship be-
`tween dose and efficacy, the drug may either be inactive or
`be active with a wide therapeutic index (Table 1).
`The information required to enable the best determination
`of dose–efficacy and dose–toxicity relationships is only
`obtainable from a randomised comparative phase II trial, as
`a single-arm trial only provides information for a single dose.
`Furthermore, unless the efficacy end-point is a response rate,
`it may not even be clear that the drug is efficacious at all, as
`demonstration of a prolonged period of stable disease may
`be due to an inadvertent selection of patients with a favour-
`able natural history, independent of any pharmacological
`drug effect.
`A further factor influencing our endorsement of random-
`ised phase II trials is the reality that a decision to move a drug
`from phase II to III is not a simple ‘thumbs up’ or ‘thumbs
`down’ decision. The standard phase II single-arm design is
`based on the promise that a difference of a single success (typ-
`ically tumour response) determines the phase III go/no-go
`decision. While even proponents of a single-arm trial likely
`realise that this is an artificial construct, the lack of randomi-
`sation in a single-arm trial hinders the ability to judge the tox-
`icity relative to control, to consider alternate end-points that
`may be more sensitive to the treatment effect, and to estimate
`the effect size relative to control that may be expected in the
`envisioned phase III trial.2 These effect sizes are the primary
`determinant of the sample size (and probability of success)
`in the phase III trial, and may also impact phase III end-point
`selection. Drugs may be active but fail in phase III because a
`phase III trial is too small, or because the chosen dose is too
`low (or high). Furthermore, it is important to have a fairly ro-
`bust estimate of the effect size, and the reliability of that esti-
`mate is determined by the sample size in phase II. Every one of
`these factors (effect size, relative toxicity, sensitive end-points
`and dose–efficacy) is better estimated through a direct ran-
`domised comparison at the phase II testing stage.
`
`Phase II
`strategy
`
`Single-arm
`trial
`
`Two-arm
`randomised
`trial with no
`control
`
`Two-arm
`randomised
`trial with
`control
`
`Multiple-arm
`randomised
`trial with
`control
`
`Result
`
`Strength of inference
`
`What are the options for design of phase II
`
`3.
`trials?
`
`Positive signal
`
`Negative signal
`
`Positive signal
`
`Negative signal
`
`Positive signal
`
`Unknown if due to true
`efficacy or bias
`Unknown if due to lack of
`efficacy, bias or wrong end-
`point
`Some sign that one agent
`more promising than
`another, but overall efficacy
`unclear
`Unknown if due to lack of
`efficacy, bias or wrong end-
`point
`Strong efficacy signal, but
`dosing may not be optimised
`
`Negative signal
`Positive signal
`
`Dose ineffective
`Strong efficacy signal, dosing
`able to be optimised
`
`Negative signal
`
`Drug ineffective
`
`Historically, phase II oncology trial designs can be divided into
`three categories: non-randomised (single-arm) trials com-
`pared to historical controls, randomised trials with multi-
`ple-experimental regimens compared to a historical control,
`and randomised trials including a prospective control arm.
`In this section, we provide a brief overview of each of these
`design strategies.
`The single-arm trial has been the most frequently used ap-
`proach to the efficacy evaluation at the phase II level in oncol-
`ogy, although it should be emphasised that this approach is
`rarely used in other therapeutic areas. Although there are
`multiple approaches to the implementation of the single-
`arm design, the overall principle has changed little since the
`design of Gehan.3 Patients are typically enrolled in two stages.
`If sufficient activity is observed at the end of the first stage, ac-
`crual continues to a second stage, and the number of suc-
`cesses (typically tumour responses) at the trial’s conclusion
`determines whether the agent is ‘recommended’ for further
`testing. Multiple variations, including optimal selection of
`
`

`
`E U R O P E A N J O U R N A L O F C A N C E R 4 5 ( 2 0 0 9 ) 2 7 5– 2 8 0
`
`277
`
`interim analysis time points, additional stages of accrual, con-
`siderations for multiple end-points, Bayesian approaches and
`approaches allowing three possible outcomes have been pro-
`posed.4–8 In an era when most new anticancer agents had very
`limited efficacy and thus any evidence of tumour activity was
`sufficient to warrant future testing, the single-arm design pro-
`vided a simple, direct and rapid method to assess an agent’s
`activity. The fundamental assumptions underlying the sin-
`gle-arm phase II trial are that data from the previous studies
`provide an adequately robust estimate of the experience the
`patients from the current trial would have had were they not
`to have received the experimental therapy, and that the end-
`point selected for the trial represents a definitive measure of
`agent activity. Both these assumptions are suspect in the cur-
`rent era. Rapidly changing standards of care in many diseases
`in therapy, imaging and supportive care imply that historical
`data may not be reflective of the results observed in the cur-
`rent clinical practice. This is compounded by the fact that ad-
`vances in tumour biology are segregating disease into marker-
`specific subtypes (e.g. based on Her-2 or K-ras status), for
`which historical data may be totally absent. In addition, new
`agents with novel mechanisms of action imply that standard
`end-points, such as tumour response, may not accurately cap-
`ture an agent’s overall patient benefit. The combination of
`these factors, in our opinion, questions the appropriateness
`of the single-arm trial at a fundamental level.
`A second class of phase II trials includes a randomisation
`to two or more experimental arms, but no prospective control
`arm. The goal of such a trial, often referred to as a selection
`design, is to provide more robust data than are available
`through single-arm trials to select the most promising exper-
`imental regimen to compare to a prospective control in the
`phase III setting. Numerous potential options for such trials
`have been proposed,9–11 all of which rely on an implicit com-
`parison to historical controls in terms of the level of activity
`required to justify moving forward the most promising exper-
`imental regimen to the phase III setting. As these phase II tri-
`als are not designed to establish a new standard of care, little
`concern is paid to the true type I error (the false-positive rate),
`which generally does not account for the multiplicity of test-
`ing (through multiple-experimental arms), as well as to an er-
`ror in the implicit historical control. While these selection
`designs do clearly provide much stronger comparative data
`to select a promising regimen amongst many possible phase
`III trials, the implicit comparison to historical controls confers
`the same disadvantages previously specified for the single-
`arm phase II trial.
`The third class of phase II trials includes a randomisation
`to one or more experimental arms and a prospective control
`arm. This is the standard approach to drug development in
`other therapeutic areas, in which the phase II programme is
`often divided into phase IIa (a two-arm trial for proof of con-
`cept) followed by phase IIb (a dose-ranging trial to optimise
`the phase III design). Two sub-classes of these randomised
`controlled designs have been used: those that formally use
`the control arm in the efficacy determination, and those that
`use the control informally. In the latter design, the control
`arm data are assessed as to its similarity to historical con-
`trols; if the control arm produces results that are ‘similar’ to
`prior experience, then the results from the experimental
`
`arm are considered valid. In our opinion, the inclusion of an
`‘informal’ control arm serves a false purpose. These trials
`are inherently comparative, and if it is recognised that the
`go/no-go decision for phase III testing will be based on many
`factors and will not be dependent on traditional statistical
`significance (p < 0.05), then to ignore the power of randomisa-
`tion to allow direct between-arm comparisons serves no valid
`scientific purpose. If the control arm is used in a formal effi-
`cacy comparison, the method of comparison is similar to that
`of a phase III trial, such as using a chi-squared comparison of
`response rates, or a log-rank comparison of progression-free
`survival (PFS) times. The formal use of the control group in
`the efficacy comparisons has been discussed by Rubinstein
`et al.,12 Fleming and Richardson13 and others.
`
`When is a non-randomised phase II trial
`4.
`sufficient?
`
`We believe non-randomised phase II trials in oncology should
`be the exception, not the rule. With that caveat, we acknowl-
`edge that such trials may be appropriate for trials in which
`the desired outcome (e.g. a partial response) will not occur
`in the absence of the investigational agent, and the rate of
`that outcome for existing agents or regimens is historically
`highly reliable. In this context, a positive phase II trial, defined
`as a response rate greater than some predefined threshold,
`would be evidence for activity, and a negative phase II trial
`would lead to the discontinuation of development for the spe-
`cific indication.
`Importantly, these criteria would never be met by many
`end-points often used in oncology clinical trials, such as ‘clin-
`ical benefit or survival beyond some threshold (unless that
`threshold was inconsistent with the natural history of the dis-
`ease). In addition, combination trials would generally not
`meet these criteria, unless the non-investigational compo-
`nents of the combination were known to be inactive in the
`target patient population. Finally, given the currently rapidly
`changing standards of care in many disease, once a new
`‘standard’ has been defined in any given disease, the histori-
`cal data obtained to date are invalid, due both to the availabil-
`ity of the new standard, and the fact that a new standard may
`change the treatment paradigm in that disease (e.g. poor
`prognosis patients who were previously untreated, thus not
`included in the historical control rates, may now become part
`of the treatment population).
`Additionally, non-randomised trials should never be used
`if there is an uncertainty regarding the optimal dose. The
`maximally tolerated dose has been traditionally utilised for
`phase II evaluation, despite the clear evidence against the
`general validity of the ‘more is better’ paradigm.14 For exam-
`ple, the randomised dose-ranging phase II trial of temsiroli-
`mus in renal cell cancer showed that doses of 25, 75 and
`250 mg weekly were all equally effective, leading to a success-
`ful phase III trial at the lowest dose.15,16
`We acknowledge that many experienced investigators
`continue to advocate the utility response rate as assessed in
`single-arm phase II trials. El-Maraghi and Eisenhauer17 re-
`viewed 89 trials of 19 agents, and concluded that the observa-
`tion of objective responses predicted for an eventual success.
`
`

`
`278
`
`E U R O P E A N J O U R N A L O F C A N C E R 4 5 ( 2 0 0 9 ) 2 7 5 – 2 8 0
`
`Given that the drugs with a 0% response rate in a single-arm
`phase II trial would not proceed to phase III testing, it is not
`surprising that no such agent achieved FDA approval. On
`the other hand, four of the six agents with the response rates
`under 10% were approved, including two agents with the re-
`sponse rates of less than 5% as single agents (cetuximab
`and sorafenib). The authors concluded that ‘even low levels
`of response may be interesting’ and acknowledged that larger
`phase II trials are required. However, the authors do not dis-
`cuss an important implication of their recommendation,
`which is likely the further increase in the number of phase
`III trials that will fail to achieve their desired end-point if all
`the drugs with the response rates of 3–4% (as observed with
`cetuximab and sorafenib) underwent phase III testing. We feel
`that PFS provides a superior signal from which to assess the
`efficacy for such agents, which is clearly assessed optimally
`through a randomised trial.
`Some authors have also suggested that the utility of sin-
`gle-arm trials using PFS or overall survival rates at a prede-
`fined landmark could be improved through the use of a
`model-based, comparison to historical controls. One example
`is a recent study by Korn and colleagues from five cooperative
`groups18 where the authors reported a multivariate prognos-
`tic model for metastatic melanoma, which they proposed
`could be utilised in single-arm phase II trials, as an alterna-
`tive to randomised trials. They suggest that only 63 patients
`would be required in a single-arm trial in this setting to have
`the same type I and II errors as a 220 patient randomised trial.
`While this model-based approach likely does provide some
`improvement compared to an unadjusted comparison to his-
`torical controls, it does not change our fundamental belief in
`the value of randomization: unless the model
`is perfect
`(which it never is), it is impossible to fully adjust the type I
`and II error for inter-trial variability, which in our opinion is
`the primary reason that single-arm phase II results are not
`replicated in phase III trials, as discussed further in the next
`section.
`
`When does a non-randomised phase II trial
`5.
`lead to a phase III trial that fails to achieve success
`on its primary end-point?
`
`As stated previously, the fundamental assumptions that
`could justify a single-arm phase II trial are that data from
`the previous studies provide an adequately robust estimate
`of the experience of the treated patients from the current trial
`were they not to have received the experimental therapy, and
`that the end-point selected for the trial represents a definitive
`measure of agent activity. Not surprisingly, when these
`assumptions are violated (which is common), phase III trials
`have a high rate of failing to demonstrate a significant
`improvement in the primary end-point from the experimen-
`tal therapy.
`From our perspective, the single greatest reason that
`uncontrolled phase II trials generate ‘false’ hope is the inter-
`trial variability in end-point success rates. Statistically, the
`failure to acknowledge the variability in the historical success
`rate inflates the nominal type I error level in the uncontrolled
`phase II trial.19 More importantly, regardless of the trial’s end-
`
`point, rapid advances in treatment outside the study protocol,
`supportive care, imaging modalities (to assess disease status)
`and other factors imply that patient outcomes will likely im-
`prove over time regardless of any new therapy. A likely even
`more dominant factor in the overall patient level success rate
`for a trial is the institutional composition of enrolling physi-
`cians and sites. Patient outcome has been repeatedly shown
`to associate with physician volume, and the patient mix at
`academic medical centres (which conduct the majority of
`phase II trials) differs widely from that in community prac-
`tice. These three factors (statistical underestimation, patient
`outcome drift and patient selection) combine to make the
`true type I error rate for the primary end-point in uncon-
`trolled phase II trials completely unknown (and unknowable).
`Multiple additional factors contribute to difficulty in pre-
`dicting successful phase III trials based on single-arm phase
`II trials. In single-arm trials of drug combinations, it is very
`difficult to distinguish the relative contribution of the stan-
`dard component from the experimental agent. The choice of
`a different end-point in a phase II trial (such as tumour re-
`sponse) compared to phase III (where overall survival or PFS
`is likely to be primary) is problematic, as response rate has
`been repeatedly shown to be a poor surrogate for these
`time-to-event end-points.
`The use of a control arm in a two-arm randomised phase II
`trial, comparing a single experimental regimen to a control,
`alleviates many of the issues above; however, several prob-
`lematic issues remain. As regimen doses and schedules are
`typically established through small phase I trials, with few
`(typically <12) patients treated at the MTD, limited informa-
`tion is available in most cases on the dose–efficacy and
`dose–toxicity relationships for the new agent. One potential
`cause of the failure in phase III trials is the error in the dose
`– either too high resulting in an unacceptable toxicity or too
`low resulting in an inadequate efficacy. This risk can be min-
`imised with a multiple-arm randomised phase II trial, where
`several dose levels are explored initially, and adaptively re-
`moved from the trial based on prespecified criteria.20
`
`What are the considerations in the design
`6.
`of randomised phase II trials?
`
`Although there has been a debate about the value of formal
`statistical comparisons in phase II trials, we feel strongly that
`such comparisons are appropriate, with the caveat that phase
`II trials do not necessarily need to provide reliable definitive
`comparisons at a traditional two-sided type I error of 0.05. Gi-
`ven that the purpose of any phase II trial is to determine
`whether to proceed with further agent development, there
`is only one outcome of interest, superiority of one or more
`experimental arms to the control. In this scenario, we believe
`a one-sided testing framework is appropriate. Given the need
`for phase II trials to be as small as possible and that a phase III
`trial will be required to confirm the efficacy in most cases,
`standard type I error rate control at the 0.05 level is not nec-
`essary. We and others12 propose that a one-sided test of the
`null hypothesis that the true primary outcome is no different
`between treatment and control with a false-positive rate of
`0.20 (type I error) is appropriate. In theory, if every phase III
`
`

`
`E U R O P E A N J O U R N A L O F C A N C E R 4 5 ( 2 0 0 9 ) 2 7 5– 2 8 0
`
`279
`
`trials were required to be justified by a randomised phase II
`trial with p < 0.20, this would lead to a phase III success rate
`of 80%, a significant improvement from the status quo. In
`contrast, in the uncontrolled phase II setting, the true false-
`positive rate is unknown.
`The use of this higher type I error remedies one common
`criticism of randomised phase II trials, that of very low statis-
`tical power.21 In the case of a multiple-experimental arm
`phase II randomised trial with a control, appropriate type I er-
`ror control is necessary to maintain an overall type I error rate
`of 20%, either through adjusting the alpha level for each com-
`parison of control versus treatment, or by comparing the
`experimental treatments to each other to identify the best,
`then comparing that regimen to control. Sample size consid-
`erations typically also dictate a type II error rate of 0.20, for a
`modest effect size (e.g. a hazard ratio of 1.5 in a time-to-event
`end-point, or a difference in response rates of 15–20%),
`although a lower type I and II error rates (e.g. 0.10) should also
`be considered if resources allow.12
`The choice of primary end-point in a randomised phase II
`trial depends on multiple factors. Ideally, a phase II screening
`trial should be a similar as possible to the subsequent phase
`III trial, including choice of primary end-point. Given the
`movement in advanced disease trials towards PFS as a phase
`III primary end-point,22 this is increasingly possible. Although
`overall survival can also be considered as the primary end-
`point for phase II and/or III testing, this would not permit
`the use of crossover designs, especially useful in the phase
`II setting.
`In our opinion, the most promising end-points for ran-
`domised phase II trials involve a comparison of a primary out-
`come measure at a single time point between the treatment
`and control groups. This outcome measure could be a tumour
`assessment at a fixed time point (e.g. change in absolute tu-
`mour size from baseline at 8–12 weeks following treatment
`initiation), or a rate of overall survival or PFS at a fixed time
`point (for example, 3 or 6 months). Such end-points facilitate
`independent radiologic review, eliminate subtle differences in
`scanning frequencies, simply patient scheduling and can be
`chosen to represent clinically meaningful time points. Ongo-
`ing research in multiple disease areas is examining these and
`other end-points to identify which end-points optimally pre-
`dict phase III success. In the future, newer methods such as
`functional imaging or assessment of circulating tumour cells
`may allow even earlier assessment of disease status.
`The validity of almost all end-points is improved by blind-
`ing, a tool infrequently used in oncology trials, particularly
`phase II trials. This is less important when overall survival
`is the primary end-point, but critical for end-points such as
`PFS, since independent radiological review can address bias
`in measurement but not bias in timing of radiological studies.
`When a trial is blinded, one can consider the use of novel end-
`points that incorporate patient-reported outcomes (e.g. time
`to symptomatic progression). In addition, blinding will result
`in a more robust analysis of toxicity attributable to the inves-
`tigational agent, as it controls for the ‘placebo effect’. Blinding
`can be logistically complicated, as it requires the availability
`of either an indistinguishable placebo (for oral agents) or
`sham parenteral dosing (for intravenous or subcutaneous
`agents). Although we feel that blinding should be considered
`
`for all randomised comparative phase II trials, we acknowl-
`edge that the final decision must consider all of these issues.
`The use of blinding also permits the use of the randomised
`discontinuation (or withdrawal) design, in which all eligible
`patients initially receive active treatment for a predefined
`run-in period. In this design, patients with stable disease at
`the end of a run-in period proceed to the randomised portion
`of the study (the primary analysis), in which they are random-
`ised to continued active treatment or placebo. To minimise
`the risk to patients randomised to the placebo arm, all pa-
`tients are offered crossover at progression, and those patients
`who had been randomised to placebo are restarted on open-
`label active treatment (those patients who had been random-
`ised to active treatment are withdrawn). This design is partic-
`ularly useful for the trials of the new agents as a single agent
`where it is hypothesised that the response rate is low, but that
`the drug has a clinically significant antitumour effect. We
`strongly feel that this design is superior in this setting to
`the use of a large single-arm trial, as a positive trial demon-
`strating a low response rate does not provide sufficient infor-
`mation to inform a robust decision to proceed to phase III
`testing.
`
`7.
`
`Conclusions
`
`Randomisation is greatly underutilised in early clinical trials
`in oncology. The use of randomised trials allows complete
`flexibility in the choice of end-points, particularly if blinding
`can be incorporated. This technique is the most powerful
`and reliable technique for distinguishing the effect of a drug
`from a placebo, a necessary predicate for a successful phase
`III trial. Given the large number of agents now available for
`clinical testing, the costs of phase III trials, and the limited
`patient and financial resources, available for such testing,
`the increased use of randomised phase II trials provides a ra-
`tional way to move new agents forward. Although end-points
`based on RECIST criteria can and should be utilised in ran-
`domised phase II trials, we do not believe that revision of
`the RECIST criteria will lead to any fundamental impact on
`improving drug development decisions in the absence of ran-
`domised clinical trials at the phase II stage of the drug
`development.
`
`Conflict of interest statement
`
`None declared.
`
`R E F E R E N C E S
`
`1. Sheiner LB. Learning versus confirming in clinical drug
`development. Clin Pharmacol Ther 1997;61(3):275–91.
`2. De Ridder F. Predicting the outcome of phase III trials using
`phase II data: a case study of clinical trial simulation in late
`stage drug development. Basic Clin Pharmacol Toxicol
`2005;96(3):235–41.
`3. Gehan EA. The determination of the number of patients
`required in a preliminary and a follow-up trial of a new
`chemotherapeutic agent. J Chronic Dis 1961;13:346–53.
`
`

`
`280
`
`E U R O P E A N J O U R N A L O F C A N C E R 4 5 ( 2 0 0 9 ) 2 7 5 – 2 8 0
`
`4. Bryant J, Day R. Incorporating toxicity considerations into the
`design of two-stage phase II clinical trials. Biometrics
`1995;51(4):1372–83.
`5. Chang MN, Therneau TM, Wieand HS, Cha SS. Designs for
`group sequential phase II clinical trials. Biometrics
`1987;43(4):865–74.
`6. Sargent DJ, Chan V, Goldberg RM. A three-outcome design for
`phase II clinical trials. Control Clin Trial 2001;22(2):117–25.
`7. Simon R. Optimal two-stage designs for phase II clinical trials.
`Control Clin Trial 1989;10(1):1–10.
`8. Thall PF, Simon R. Practical Bayesian guidelines for phase IIB
`clinical trials. Biometrics 1994;50(2):337–49.
`9. Sargent DJ, Goldberg RM. A flexible design for multiple armed
`screening trials. Stat Med 2001;20(7):1051–60.
`10. Schaid DJ, Ingle JN, Wieand S, Ahmann DL. A design for phase
`II testing of anticancer agents within a phase III clinical trial.
`Control Clin Trial 1988;9(2):107–18.
`11. Simon R, Thall PF, Ellenberg SS. New designs for the selection
`of treatments to be tested in randomized clinical trials. Stat
`Med 1994;13(5–7):417–29.
`12. Rubinstein LV, Korn EL, Freidlin B, Hunsberger S, Ivy SP, Smith
`MA. Design issues of randomized phase II trials and a
`proposal for phase II screening trials. J Clin Oncol
`2005;23(28):7199–206.
`13. Fleming TR, Richardson BA. Some design issues in trials of
`microbicides for the prevention of HIV infection. J Infect Dis
`2004;190(4):666–74.
`14. Winer EP, Berry DA, Woolf S, et al. Failure of higher-dose
`paclitaxel to improve outcome in patients with metastatic
`
`breast cancer: cancer and leukemia group B trial 9342. J Clin
`Oncol 2004;22(11):2061–8.
`15. Atkins MB, Hidalgo M, Stadler WM, et al. Randomized phase
`II study of multiple dose levels of CCI-779, a novel
`mammalian target of rapamycin kinase inhibitor, in patients
`with advanced refractory renal cell carcinoma. J Clin Oncol
`2004;22(5):909–18.
`16. Hudes G, Carducci M, Tomczak P, et al. Temsirolimus,
`interferon alfa, or both for advanced renal-cell carcinoma. N
`Engl J Med 2007;356(22):2271–81.
`17. El-Maraghi RH, Eisenhauer EA. Review of phase II trial designs
`used in studies of molecular targeted agents: outcomes and
`predictors of success in phase III. J Clin Oncol
`2008;26(8):1346–54.
`18. Korn EL, Liu PY, Lee SJ, et al. Meta-analysis of phase II
`cooperative group trial

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