throbber
COMMENTARY
`
`Statistical and Ethical Issues in the Design
`and Conduct of Phase I and II Clinical
`
`Trials of New Anticancer Agents
`
`Mark J. Ratain, Rosemarie Mick, Richard L. Schilsky,
`Mark Siegler*
`
`The development of new anticancer agents is a complex,
`stepwise process proceeding from discovery to demonstra-
`tion of
`antitumor
`activity
`in
`preclinical models
`and
`evaluation of nonnal
`tissue toxicity prior to initiation of
`clinical trials. The purpose of the initial clinical trials (phase
`I)
`is
`to define the toxic effects of
`the agent and the
`recommended dosage for subsequent
`testing (1). The vast
`majority of drugs that reach phase I studies go on to phase II
`testing, which is aimed at finding evidence of efficacy in
`human cancer
`(1). Rarely, drugs
`are withdrawn at
`the
`completion of phase I
`testing, usually due to unpredictable
`and/or severe nonhematologic toxic effects that may be
`irreversible (2-4).
`Although there is general agreement regarding the overall
`research goals of phase I and phase II
`studies of new
`anticancer agents,
`two problems emerge:
`I) The research
`goals may differ from the patient’s goals, and 2) there is no
`consensus on how to achieve the researcher’s goals in the
`most
`efficient
`and ethically appropriate way.
`In
`this
`commentary, we discuss both statistical and ethical issues of
`early anticancer drug development and suggest an alternative
`framework that may improve the protocol design and address
`the ethical
`issues of phase I and phase II clinical
`trials.
`
`Phase I Trials: Statistical Issues
`
`Background
`
`is to
`trial
`The major scientific objective of the phase I
`determine the recommended phase II dose of the drug being
`studied. Thus, phase I trials address an estimation problem
`rather than the testing of a hypothesis. The theoretically
`optimal dose for any individual patient is the dose resulting
`in the maximally achievable antitumor response with an ac-
`ceptable level of toxicity. Since one cannot predict efficacy
`prior to treatment,
`the theoretically optimal
`individual dose
`is the maximal dose that does not exceed an acceptable level
`
`are designed to
`trials
`I
`toxicity. However, phase
`of
`determine the recommended phase II dose for a population,
`not an individual. Thus, since there is usually substantial
`interpatient variability in toxic effects,
`the recommended
`phase II dose will always be an imprecise estimate of the
`optimal dose for the individual patient (5). As a result, many
`phase II trials utilize intrapatient dose modifications based
`on observed toxicity in an attempt to treat each patient at the
`optimal dose.
`A second problem with the design of phase I trials is that
`the optimal dose is usually administered to only a minority
`of the patients treated in the phase I trial. The reason for this
`occurrence is complicated. For detemiination of the recom-
`mended phase II dose, it is standard research practice to treat
`cohorts of patients beginning at a dose that is believed to be
`nontoxic
`and then escalating the dose for patients
`in
`successive cohorts until defined grades of toxic effects are
`observed. Table l
`lists the variables defined by the in-
`vestigator who uses the traditional phase I design and some
`of the commonly considered alternatives.
`Statistical issues in phase I trials were initially outlined by
`Schneiderman (6). Other
`investigators have subsequently
`considered such issues with regard to the traditional design,
`such as the optimal starting dose (7-10) and whether real-
`time pharmacokinetics can more quickly lead to dose escala-
`tions
`(11-13). However,
`investigators have not carefully
`focused on criteria for tennination of phase I studies, which
`should only occur when the recommended phase II dose has
`been adequately defined.
`If all
`three patients of a hypothetical cohort experience
`dose-limiting toxicity, most
`investigators would agree that
`the recommended phase II dose has been exceeded. How-
`ever, even in that setting, the investigator must acknowledge
`that the 95% exact confidence interval for the incidence of
`
`‘See “Notes" section following “Rcfercnccs."
`
`Journal of the National Cancer Institute, Vol. 85. No. 20, October 20, 1993
`
`COMMENTARY 1637
`
`Amerigen Exhibit 1102
`Amerigen Exhibit 1102
`Amerigen v. Janssen IPR2016-00286
`Amerigen V. Janssen IPR20l 6-00286
`
`

`
`
`
`Table 1. Variables to be defined for traditionally designed phase I trials‘
`
`
`Variable
`
`Staning dose
`Patients per nontoxic dose level
`Definition of DLT
`Subsequent dose levels
`Patients per toxic dose level
`Definition of RPTD
`Patients at RPTD
`
`Example
`
`‘/io LD,0
`3
`Any grade 3 toxicity
`“Modified Fibonacci"
`6
`<2 of 6 patients with DLT
`6
`
`Suggested alternatives
`
`Higher
`Fewer
`Grade 3-41’
`Pharmacokinetically guided dosing
`Greater
`<3 of 6 patients with DLT
`Much greater
`
`‘DLT = dose-limiting toxicity; RPTD = recommended phase II dose.
`TGrade 3 organ toxicity is dose-limiting, but grade 3 myelosuppression or nausea/Vomiting is not.
`
`rate of
`If the target
`dose-limiting toxicity is 37%—l00%.
`dose-limiting toxicity is 50%, then the recommended phase
`H dose may be underestimated. Many investigators will not
`accept a 33% rate of dose-limiting toxicity (two of six
`patients) at the recommended phase II dose. Yet,
`the 95%
`exact confidence interval for this dose-limiting toxicity rate
`is quite broad (6%-73%).
`
`Alternatives to “Standard” Phase I Trial Design
`
`Because of the relatively small number of patients who
`actually receive the recommended phase II dose, several
`authors (5,14-22) have suggested substantial modifications to
`the traditional phase I trial design. These modifications have
`been aimed at efficiently determining the recommended
`phase II dose, while minimizing the risk of both undertreat-
`ment and excessive toxicity. Since most responses in phase I
`studies occur at dose levels that are 80%-120% of the
`
`recommended phase II dose (23), such modified trial designs
`would result in a greater response rate during phase I. For
`example, several
`investigators (14.16) have suggested “up
`and down” designs,
`in which the dose for each patient
`is
`assigned on the basis of the experience in the patient(s) most
`recently treated and may be adjusted either higher or lower.
`These designs are relatively simple modifications to the
`traditional design, but they base major protocol decisions on
`relatively small numbers of patients.
`In contrast
`to the traditional design and these modifica-
`tions of it, O’Quigley et al.
`(17,18) proposed a Bayesian
`approach,
`the continual
`reassessment method. A Bayesian
`method begins with assumptions about the main end points
`of the study, known as “priors,” which are derived from the
`investigators’ prior observations and/or beliefs based on their
`own experience and that of others.
`In a phase I trial
`that
`uses
`a Bayesian approach,
`infonnation 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 H dose. Patients are then treated
`at
`the current estimated recommended phase II dose, and
`these
`estimates
`are
`continually updated to reflect
`the
`accumulating dose—toxicity data. When the sample size
`(determined in advance) has been reached, the final estimate
`of the recommended phase II dose is made from all available
`data. Although it
`is desirable to meet
`the ethical goal of
`treating each patient at the hypothesized recommended phase
`
`II dose, there is a danger that patients would be exposed to
`highly toxic doses
`if major errors were made
`in
`the
`assumptions about
`the end points of the study.
`is a bridge
`We recently proposed a design (22)
`that
`between the traditional design and the Bayesian design
`suggested by O’Quigley et al. (17,18). The scheme utilizes a
`cohort-based escalation approach similar
`to that used in
`traditional phase
`I
`studies. As dose—toxicity data
`are
`accumulated, a pharmacodynamic model
`is fit
`to the data.
`Model-guided dosing commences only after a prespecified
`number of patient cohorts have been treated and evaluated
`for toxic effects and after a dose—toxicity relationship is
`statistically defined. Thus, model-guided dosing is
`less
`dependent on assumptions and more dependent on observed
`toxic effects. Unlike the design proposed by O‘Quigley et al.
`(17,18),
`each patient
`is not
`treated at
`the
`estimated
`11
`recommended phase
`close, but
`instead at
`the dose
`calculated to yield a target nadir white blood cell count for
`the individual patient. This design is only useful when
`myelosuppression is dose limiting, although modifications to
`include graded nonhematologic toxicity would be feasible.
`Based on computer simulation studies, this method performs
`better than the traditional design because it requires entry of
`fewer patients
`in the phase I
`study and yields more
`reproducible estimates of the recommended phase II dose for
`an average patient. As pointed out by Mathe and Brienza
`(24),
`there may be substantial
`interstudy variability in the
`recommended phase II dose.
`
`Phase I Trials: Ethical Issues
`
`Background
`
`Investigators conducting phase I trials must adhere to the
`ethical norms of clinical
`research (25-30) and therefore
`encounter a number of potential ethical
`issues:
`1) Minimizing patients treated at
`ineffective doses;
`2) Minimizing patients treated at
`toxic doses;
`3) Historically low probability of response in phase I
`trials;
`
`4) Unknown toxicity and benefit of new agent; and
`5) Difficulty
`in obtaining true
`informed consent
`vulnerable patient populations.
`Some have argued that since phase I nials are designed to
`define the qualitative and quantitative aspects of toxicity,
`
`in
`
`1638 COMMENTARY
`
`Journal of the National Cancer Institute, Vol. 85, No. 20, October 20,
`
`l993
`
`

`
`patients should only enter such trials “for the benefit of
`future cancer victims” (26). While this situation may apply
`to normal volunteers without disease who enroll
`in phase I
`trials of some drugs, it surely does not apply to phase I trials
`of anticancer agents, since both the control and treatment
`arms of the trial only involve patients with cancer (31).
`Thus, cancer treatments in phase I
`trials are always ad-
`ministered with therapeutic intent, even though the major
`scientific objective of a phase I study is to define toxicity.
`Most importantly, the ethical concerns related to therapy for
`patients with advanced cancer do not depend on whether or
`not the treatment is administered in the context of a clinical
`
`trial or whether such a trial is phase I, phase II, or phase HI.
`
`Risk—Benefit Ratio
`
`Patients offered treatment in the context of a phase I trial
`are informed that
`the purpose of the study is to find the
`optimal drug dose and that some patients will be treated at a
`dose that
`is too low or too high (i.e.,
`too toxic). They are
`also informed that
`there is a small possibility that
`the
`treatment will be beneficial. The probability of benefit,
`however, does not depend on the type of trial, but rather on
`the disease being treated, the drug being tested, and the dose
`of the drug that
`is administered. For example,
`there is
`probably a greater response rate for lymphomas in phase I
`trials than for pancreatic cancer in phase III trials, since the
`latter is rarely responsive to any therapy.
`Phase I studies often raise ethical concerns because of the
`
`is exposed to drug toxicity
`the patient
`perception that
`without being offered a reasonable expectation of benefit
`(30). Another common concern is the potential for under-
`treatment of those patients enrolled at dose levels that are
`eventually determined to be subtherapeutic. These concerns
`have led some to suggest
`the use of
`intrapatient dose
`escalation as a solution to this problem (24). With this
`approach, patients who experience little or no toxicity at
`their entry dose receive increased doses on subsequent
`cycles. However, intrapatient dose escalation only allows for
`the possibility that patients who do not rapidly succumb to
`their disease might eventually receive a “therapeutic” dose
`of the new agent. This approach does not recover the time
`lost while the patient received ineffective therapy, and it
`does not clearly increase the chance of tumor response, since
`the tumor is likely to be resistant even to doses that result in
`toxic
`effects
`unless
`such
`dose
`escalation
`is marked
`
`(>100%). In addition, it increases the period of time that a
`patient
`remains on a particular
`study, possibly limiting
`opportunities for treatment in subsequent trials. Traditionally
`designed phase I studies do have a unique property: At least
`one patient must experience dose-limiting toxicity for the
`trial
`to be completed. A statement
`to this effect
`is now
`included in the consent fonn of phase I cancer trials at our
`institution.
`
`substantial and rewarding response, as was observed during
`the phase I
`trials of cisplatin (33) and paclitaxel
`(Taxol)
`(34).
`In fact, all 17 drugs that were eventually marketed
`after beginning National Cancer Institute-sponsored phase I
`testing from 1970 through 1983 manifested activity in at
`least one phase I
`trial
`(22). Thus,
`the potential range of
`outcomes, both beneficial and harmful,
`is extremely broad.
`At study initiation, the type and degree of toxicity likely
`to occur are unknown, although toxicology studies are often
`predictive for drug toxicity to organs. Patients must
`take
`risks
`that cannot be
`easily measured. Since both the
`probability of benefit and toxicity are unknown, estimating
`the therapeutic index is impossible. At low dose levels, there
`is a low probability of both toxicity and benefit. At dose
`levels near the recommended phase H dose, there is a high
`probability of severe toxicity and a maximal probability of
`benefit
`(23). The magnitude of the probability of benefit
`cannot be ascertained until phase II
`trials have been
`completed.
`
`Informed Consent
`
`An issue of great concern to both investigators and
`institutional
`review boards
`is
`the
`consent process
`(27,29,30,35,36), with the suggestion by some of procedural
`safeguards
`such as
`third-party consultation with nonin-
`vestigators (i.e., primary care physicians) as patient advo-
`cates (30). The major obstacle to true “informed consent” is
`that
`the investigator often has little information. Consent
`forms usually contain a litany of possible side effects, when
`in fact the clinical experience has been almost exclusively
`without toxicity. Furthermore, centers with a major interest
`in phase I trials may have 10 or more trials available to an
`individual patient. Should patients be offered all such trials
`as alternatives? Since there may be substantial deficiencies
`in participants’ perceptions of the consent process in even
`less complex trials (36), subjects may be confused regarding
`what
`is involved in any specific trial.
`Freedman (35) has suggested a cohort—specific approach to
`phase I cancer
`studies.
`In this approach,
`the informed
`consent process is different for the first patient entered in the
`study compared with that used to enroll patients after
`significant toxicity has been noted. This strategy requires a
`dynamic consent form, which varies according to the patient
`cohort. One expects
`that
`investigators would ordinarily
`communicate this information to the patient verbally, but
`inclusion of this information as an appendix to the standard
`consent form would ensure that
`the patient is informed of
`the actual experience to date.
`
`Phase II Trials: Statistical Issues
`
`Background
`
`the low
`is
`trials
`Another major concern in phase I
`probability of response. There is no question that, on the
`basis of bidimensional
`tumor measurements, most patients
`entered in phase I trials do not achieve a partial or complete
`response (32). However,
`there is the small possibility of a
`
`Phase II trials are generally studies with no control group
`that are aimed at estimating the antitumor efficacy of a new
`agent in a particular disease. The design of such trials has
`been discussed extensively in the literature;
`the discussion
`has recently focused on issues of sample size and hypothesis
`
`Joumal of the National Cancer Institute, Vol. 85, No. 20, October 20, I993
`
`COMMENTARY 1639
`
`

`
`testing as a basis for guidelines relating to early discontinua-
`tion of therapy if there is adequate evidence of inefficacy
`(15,37—51).
`a
`test
`trials
`trials, phase II
`to phase I
`In contrast
`hypothesis: “New drug X is active against disease Y.” The
`investigator must still define “active” as well as select
`which subset of patients with disease Y will be studied. For
`example,
`the investigator may wish to determine whether
`drug X has a 20% response rate in women with metastatic
`breast cancer
`refractory to doxorubicin.
`In the simplest
`design, based on binomial probability, 14 patients would be
`evaluated initially. If no patient responds,
`the investigator
`may conclude with 95% certainty that drug X has a response
`rate less than 20% in this patient population. If at least one
`patient
`responds,
`a second cohort of patients would be
`treated to better estimate the response rate. The greater the
`number of patients in this second cohort, the more precisely
`one can estimate the response rate.
`
`Alternative Designs
`
`to this design have been
`A number of variations
`suggested. Fleming (43) proposed testing two hypotheses
`simultaneously, one for a minimally acceptable response rate
`and one for a maximally unacceptable response rate (i.e., the
`highest response rate an investigator would be willing to
`miss). Simon et al.
`(45) suggested randomized phase II
`trials. This design is similar to the Fleming design in that it
`tests two (or more) hypotheses simultaneously: a) drug A is
`active against disease Y, and b) drug B is active against
`disease Y.
`
`End Points
`
`the
`Virtually all phase II trials use a common end point,
`objective response rate, which is defined as the percentage
`of assessable subjects who demonstrate a partial or complete
`response. Is screening new agents for a l5%-20% objective
`response rate the best approach? One could argue that we
`are using needlessly strict criteria and that we should simply
`be looking for improvement in quality of life or decrease in
`the rate of tumor growth, possibly by utilizing sequential
`bidimensional measurements (52). Clearly,
`the end point
`should be based on the patient population to be studied.
`
`Phase II Trials: Ethical Issues
`
`Probability of Response
`
`lung cancer. In a separate analysis (54),
`for non—small—cell
`the overall response rate for new agents in phase II trials in
`non—small—cell
`lung cancer was 4%, which is less than the
`overall objective response rate in phase I
`trials of 6%
`reported by Von Hoff and Turner
`(23). Thus, one can
`conclude that
`the vast majority of patients with advanced
`solid tumors that are either refractory to standard therapy or
`for which no standard therapy exists will not respond to any
`treatment, whether
`it be noninvestigational, phase II, or
`phase I.
`
`Informed Consent
`
`trials is the
`issue associated with phase II
`One ethical
`construct used for hypothesis testing,
`i.e., attempting to
`disprove inactivity. The null hypothesis being tested in phase
`II
`trials is
`that “Drug X has less than a Z% objective
`response rate in disease Y.” Thus, responses disprove the
`investigator’s null hypothesis, and, usually, an expansion of
`the trial
`is then required to confirm the response rate. But
`how do investigators view their evolving data from phase II
`trials? What do we tell the 14th patient if all of the prior 13
`have failed to respond? Do we say, “If you don’t respond,
`we will be 95% certain that drug X has less than a 20%
`objective response rate in disease Y”?
`Sordillo and Schaffner (55) have previously addressed this
`issue and have recommended that a statement be added to
`
`the consent form regarding the lack of activity to date. But
`do we really need the 14th patient? The investigator has
`already concluded, albeit with only 94% certainty, that drug
`X is inactive (<20% objective response rate) in disease Y.
`But
`if we accept 13 patients as the final sample size,
`then
`what do we tell
`the 13th patient
`if none of the first 12
`responded? This recursive reasoning can be iterated back to
`the beginning of the study!
`
`Vulnerability of Subjects
`
`Patients entering phase II trials are as vulnerable as those
`entering phase I trials. Whereas patients may often select a
`phase I
`trial
`from multiple studies available at a major
`center, investigators have been cautioned against conducting
`more than one phase II trial for the same patient population,
`because of the risk of bias in patient selection in individual
`studies. Thus, patients offered a phase II trial at a particular
`center may feel
`that
`they have
`no good altematives,
`regardless of the data accrued to date.
`
`Patient Selection
`
`trials have received little
`in phase II
`issues
`Ethical
`attention in the past. Whereas it
`is commonly known that
`very few patients achieve partial or complete responses
`during phase I studies (32), little concern has been expressed
`among investigators regarding the generally poor results of
`phase II trials. Marsoni et al. (53) tabulated the results of all
`National Cancer Institute-sponsored phase II trials from 1970
`to 1985 and found a single active drug from among 42 phase
`II trials for colon cancer and from among 33 phase II trials
`
`recognize the problems with current
`investigators
`As
`phase II designs, new ethical
`issues must be addressed.
`Currently, an important
`issue under debate is whether to
`include in phase II trials previously untreated patients with
`tumors that are sensitive to chemotherapy but incurable, such
`as metastatic breast cancer or extensive small-cell
`lung
`cancer (56-61). This issue was recently considered in detail
`by Moore and Korn (62).
`
`1640 COMMENTARY
`
`Jouma] of the National Cancer Institute, Vol. 85, No. 20, October 20, 1993
`
`
`
`

`
`Comparison of Ethical Issues for Phase I and
`Phase II Trials
`
`trials place the investigator/
`Both phase I and phase II
`physician under “ethical stress” (26); in both types of trials,
`the statistical and clinical objectives are in conflict. The
`conflict
`in phase I between the scientific goals of defining
`toxicity,
`recommended dosage, and clinical pharmacology
`and the therapeutic goal of antitumor effect
`is obvious to
`many and is generally recognized by both investigators and
`Institutional Review Board chairs (30). The conflict in phase
`II is less obvious, depending on the extent of accrual to the
`study and the results in the assessable patients to date (55).
`Patients may enroll
`themselves
`in
`clinical
`trials
`for
`altruistic reasons, but most do so primarily in the hope of
`therapeutic benefit (3036). Patients seek the benefit but may
`in turn benefit
`from the hope itself. Those patients who
`enroll
`in phase I
`trials are usually aware of the lack of
`knowledge of efficacy regarding drug X at dose Z in disease
`Y. However, one must also be concerned about the consent
`
`process in phase II trials. It is not clear that patients are told
`that “drug X has been shown to be safe, but there are no
`data yet to suggest that it is effective in disease Y.” Even if
`there are no clinical data, the patient might feel that drug X
`is likely to be effective. It is even less clear that the patient
`will be informed of negative data as the trial evolves.
`The decision to enroll in a clinical trial should include an
`
`assessment of the probability of both incremental benefit and
`harm. A major difference between phase I and phase H trials
`is
`in the assessment of toxicity.
`It may be paradoxical
`to
`some that most patients in phase I trials experience less drug
`toxicity than those in phase II
`trials. This paradox occurs
`because most patients in phase I studies are undertreated
`(23.30), whereas patients in phase II
`trials are generally
`treated at a dose that results in moderate to severe toxicity.
`Thus, on the average, toxicity in phase I studies is less than
`in phase II, but there is a much greater degree of variation.
`Most patients in phase I studies have a low probability of
`both benefit and toxicity. Their major costs are the in-
`convenience of being in the study and “opportunity risk” if
`there are other possible treatments. In contrast, most patients
`in phase II
`trials have a significant risk of experiencing
`toxicity and an unknown likelihood of benefit, at
`least at
`study initiation. From the standpoint of the patient and
`treating physician, the late stages of a phase I trial, in which
`the dose being used is near the recommended phase II dose,
`are virtually equivalent to the early stages of a phase II trial,
`because the dose and toxicity have been identified and the
`benefit
`is unknown.
`
`Considerations for the Future
`
`Phase I Trials
`
`The traditional cohort design is clearly imperfect, and it is
`important to consider implementation of new study designs,
`such as the design proposed by O’Quigley et al. (17,I8) or
`the model-based dosing design that we have suggested (22).
`Phase
`I
`studies
`should attempt
`to define more
`than
`
`emphasizing instead the
`recommended phase H dose,
`development of a preliminary pharmacodynamic model to be
`tested prospectively in phase II
`trials (63), particularly in
`conjunction with limited sampling strategies based on prior
`pharmacokinetic studies (64-66).
`How much patient autonomy should there be in phase I
`studies? The opportunity for a patient
`to choose among
`multiple phase I
`trials is one example. A more difficult
`autonomy issue is whether patients should have a say in the
`dose of the drug they wish to receive.
`If the investigator
`does not know the optimal dose, might it be reasonable to
`treat patients willing to take greater risks with higher doses?
`Suppose 10 dose levels have been defined for drug X,
`beginning at the standard starting dose (one-tenth of the dose
`lethal
`to 10% of treated animals). Patients might be given
`the choice of receiving the current dose level N, which has
`been incompletely evaluated, or dose level N + 1, which has
`not been evaluated. If a patient selected level N + 1 and
`experienced no toxicity,
`level N could be dropped. Then,
`patients would choose between level N + 1 and level N + 2.
`This approach could be further expanded to allow patients
`even greater autonomy, such as being allowed to choose
`level N + 2 or even N + 8. Thus, more aggressive patients
`could expedite completion of a study, assuming that
`the
`consent process can be completed in a fully infonned,
`ethical
`setting. An important concern, however,
`is
`the
`patient’s
`ability to make
`such difficult
`and
`complex
`decisions. Clearly,
`this would require a dynamic consent
`fonn (35) and probably third—party consultation with a
`noninvestigator as patient advocate.
`
`Phase II Trials
`
`the ethical
`Investigators must become more aware of
`issues in phase II trials. A dynamic consent process could be
`implemented to inform patients of results available to date,
`particularly when there is a high probability that the drug is
`ineffective (55). This may make single-institution phase II
`trials more difficult to complete unless patients are accrued
`rapidly. It would also require better communication among
`those institutions participating in multi—institution studies.
`Phase II trials involving multiple institutions and multiple
`drugs (tested as single agents) would be a possible solution
`to this
`issue.
`In order to maximize patient autonomy, a
`randomized
`consent
`design could
`be
`used
`(67),
`or,
`alternatively, the patient and treating physician could jointly
`select an agent on the basis of data accrued to date. Since
`the purpose of phase II is to decide whether further testing is
`warranted, this self—selection by patients would not necessar-
`ily alter conclusions
`(55). Patients would be
`likely to
`“follow the winner” once activity is
`identified.
`If this
`activity is not confirmed, subsequent patients would be less
`likely to select
`the agent.
`In addition, patients would be
`guided by the type and degree of toxicity to be expected.
`The major drawback to such innovative designs, which
`require a high level of patient participation,
`is the concern
`that the complexity of the consent process could lead to the
`exclusion of
`those patients
`intellectually or emotionally
`unable to participate.
`
`Joumal of the National Cancer Institute, Vol. 85, No. 20, October 20, 1993
`
`COMMENTARY 1641
`
`

`
`
`
`We must also reconsider the value of demonstrable data
`
`than a partial
`less
`is
`that
`regarding therapeutic benefit
`should be considered
`response. Palliation of
`symptoms
`highly important, and new agents with little toxicity and
`documented symptomatic benefit might warrant approval
`with a relatively low objective response rate. For example,
`the potential usefulness of a drug with 15 minor responses
`associated with symptomatic benefit
`in 18 patients is very
`different from that of a drug with no responses at all, yet
`both might have an objective response rate of 0%. The
`determination of a minimum target objective response rate
`depends on the clinical setting; a 20% objective response
`rate in primary pancreatic cancer is much more exciting than
`a 20% objective response rate in untreated metastatic breast
`cancer. Since evidence of activity in phase I has
`some
`predictive value (23). this infonriation could also be utilized
`in the implementation of new phase 11 designs (48).
`A more innovative approach to phase I and phase II
`clinical
`trials
`is
`important. We must expand beyond a
`hypothesis-testing framework driven by a defined P value
`(68,69). Biostatisticians and clinicians should both consider
`
`phase I evaluation as an ongoing process by which a dose-
`toxicity relationship is refined and understood. Implementa-
`tion of
`frequent data evaluation with proper Bayesian
`analysis
`(19,22,48,68,70,7I) will allow us
`to efficiently
`evaluate new agents and preserve our patients’ dignity and
`autonomy in the process.
`
`References
`
`IJ. et al: EORTC New Drug
`(1) Schwartsmann G, Wanders J. Koier
`Development Office coordinating and monitoring programme for
`phase I and 11
`trials with new anticancer agents. Eur
`J Cancer
`272ll62—1l68, 1991
`trial of
`(2) Brown TD, Ettinger DS, Donehower RC: A phase I
`spirohydantoin mustard (NSC 172112)
`in patients with advanced
`cancer. J Clin Oncol 4:1270—1276. 1986
`(3) Margolin K. Doroshow J, Leong L, et al: 3-Deazaguanine: report of a
`phase I
`trial and drug-related cardiac toxicity.
`Invest New Drugs
`81369-376. 1990
`(4) Schilsky RL, Ratain MJ, Janisch L, et al: Phase I clinical and
`pharmacologic study of 502U83 administered as a 24-hour continuous
`intravenous infusion. Cancer Chemother Pharmacol 31:283-288. 1993
`(5) Edler L: Statistical requirements of phase I studies. Onkologie 13:90-
`95, 1990
`(6) Schneidennan MA: Mouse to man: statistical problems in bringing a
`drug to clinical trial. In Proceedings of the 5th Berkeley Symposium
`on Mathematical Statistics and Probability, vol. 4. Berkeley: Univ
`Calif Press, 1967. pp 855-866
`(7) Freireich EJ, Gehan EA. Rall DP. et al: Quantitative comparison of
`toxicity of anticancer agents in mouse. rat. hamster. dog, monkey. and
`man. Cancer Chemother Rep 50:219-244. 1966
`(8) Penta JS, Rozencweig M, Guarino AM, et al: Mouse and large-animal
`toxicology studies of twelve antitumor agents: relevance to starting
`dose for Phase I clinical
`trials. Cancer Chemother Pharmacol 3:97-
`101. 1979
`toxicology to
`(9) Grieshaber CK, Marsoni S: Relation of preclinical
`findings in early clinical trials. Cancer Treat Rep 70:65-72. 1986
`Penta JS. Rosner GL, Trump DL: Choice of starting dose and
`escalation for phase 1 studies of antitumor agents. Cancer Chemother
`Pharmacol 312247-250, 1992
`for
`roles
`(ll) Collins JM, Laharko DS, Dedriclr RL, et al: Potential
`preclinical pharmacology in phase 1 clinical
`trials. Cancer Treat Rep
`70:73-80, 1986
`trials.
`(12) Pharmacokinetically guided dose escalation in phase I clinical
`Commentary and proposed guidelines. EORTC Pharmacokinetics and
`Metabolism Group. Eur J Cancer Clin Oncol 211083-1087. 1987
`
`(10)
`
`(13) Collins JM, Grieshaber CK, Chabner BA: Pharmacologically guided
`phase I clinical trials based upon preclinical drug development. J Natl
`Cancer Inst 8221321-1326. 1990
`(14) Bolognese JA: A Monte Carlo comparison of three up-and-down
`designs for dose ranging. Controlled Clin Trials 42187-196, 1983
`(15) Geller NL: Design of phase I and II clinical
`trials in cancer: a
`statistician's view. Cancer Invest 2:483—491, 1984
`(I6) Storer BE: Design and analysis of phase I clinical trials. Biometrics
`45:925-937. 1989
`(17) O'Quigley J, Pepe M, Fisher L: Continual reassessment method: a
`practical design for phase I clinical trials in cancer. Biometrics 46:33-
`48. 1990
`(I8) O'Quigley J, Chevret S: Methods for dose finding studies in cancer
`clinical trials: a review and results of a Monte Carlo study. Stat Med
`10:l647—1664. 1991
`(19) Gatsonis C, Greenhouse JB: Bayesian methods for phase I clinical
`trials. Stat Med ll:l377—l389. 1992
`(20) Gordon NH. Willson JK: Using toxicity grades in the de

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket