throbber
REVIEW
`
` Dose Escalation Methods in Phase I Cancer Clinical Trials
` Christophe Le Tourneau , J. Jack Lee , Lillian L. Siu
`
` Phase I clinical trials are an essential step in the development of anticancer drugs. The main goal of these studies is to establish the
`recommended dose and/or schedule of new drugs or drug combinations for phase II trials. The guiding principle for dose escalation
`in phase I trials is to avoid exposing too many patients to subtherapeutic doses while preserving safety and maintaining rapid
`accrual. Here we review dose escalation methods for phase I trials, including the rule-based and model-based dose escalation meth-
`ods that have been developed to evaluate new anticancer agents. Toxicity has traditionally been the primary endpoint for phase I
`trials involving cytotoxic agents. However, with the emergence of molecularly targeted anticancer agents, potential alternative end-
`points to delineate optimal biological activity, such as plasma drug concentration and target inhibition in tumor or surrogate tissues,
`have been proposed along with new trial designs. We also describe specific methods for drug combinations as well as methods
`that use a time-to-event endpoint or both toxicity and efficacy as endpoints. Finally, we present the advantages and drawbacks of
`the various dose escalation methods and discuss specific applications of the methods in developmental oncotherapeutics.
`
` J Natl Cancer Inst 2009;101: 708 – 720
`
` Phase I trials represent the first application of a new drug or drug
`combination to humans and as such are the foundation of a success-
`ful clinical drug development process. Because the early clinical
`development of a novel agent may unduly influence its ultimate fate,
`a careful and thoughtful approach to the design of phase I trials is
`essential. Phase I clinical trials in oncology are typically small, sin-
`gle-arm, open-label, sequential studies that include patients with a
`good performance status whose cancers have progressed despite
`standard treatments. A principal goal of such trials is to establish the
`recommended dose and/or schedule of an experimental drug or
`drug combination for efficacy testing in phase II trials. A phase I trial
`design has many components, including starting dose, dose incre-
`ment, dose escalation method, number of patients per dose level,
`specification of dose-limiting toxicity, target toxicity level, definition
`of the maximum tolerated dose (MTD) and recommended dose for
`phase II trials, patient selection, and number of participating centers
`( see definitions of basic concepts in Table 1 ). Although all of these
`components are relevant for the design of a phase I trial, this review
`will focus on selecting the dose escalation method that will yield an
`optimal balance of safety, efficiency, and ethical conduct.
` The guiding principle for dose escalation in phase I trials is to
`avoid unnecessary exposure of patients to subtherapeutic doses of
`an agent (ie, to treat as many patients as possible within the thera-
`peutic dose range) while preserving safety and maintaining rapid
`accrual. Dose escalation methods for phase I cancer clinical trials
`fall into two broad classes: the rule-based designs, which include
`the traditional 3+3 design and its variations, and the model-based
`designs. The rule-based designs assign patients to dose levels
`according to prespecifi ed rules based on actual observations of
`target events (eg, the dose-limiting toxicity) from the clinical data.
`Typically, the MTD or recommended dose for phase II trials is
`determined by the prespecifi ed rules as well. On the other hand,
`the model-based designs assign patients to dose levels and defi ne
`the recommended dose for phase II trials based on the estimation
`of the target toxicity level by a model depicting the dose – toxicity
`
`relationship. However, because of safety concerns, most model-
`based designs are modifi ed such that specifi c restrictions are set as
`safeguards for elements such as dose increments to avoid over-
`shooting of the MTD and thus exposing patients to undue harm.
`All of these methods were developed in the era of cytotoxic drugs,
`during which time it was assumed that both effi cacy and toxicity
`increase with dose. These relationships are typically represented
`by dose – toxicity and dose – effi cacy curves in which toxicity and
`effi cacy increase monotonically with increasing dose ( Table 1 and
` Figure 1 ). Consequently, these methods have used toxicity as the
`primary endpoint. For molecularly targeted agents, the dose –
`effi cacy and dose – toxicity curves may differ from those for cyto-
`toxic agents, and effi cacy may occur at doses that do not induce
`clinically signifi cant toxicity ( 1 – 4 ). Thus, for trials involving these
`agents, the occurrence of drug-related biological effects has been
`suggested as an alternate primary endpoint besides toxicity ( 1 – 4 ).
` Here we review the different dose escalation methods for phase I
`cancer clinical trials of single agents and drug combinations and
`discuss their pros and cons. Recent reviews ( 2 , 5 ) of phase I clinical
`trials including this update reveal that new dose escalation designs
`have been incorporated into phase I trials infrequently, and we
`
` Affiliations of authors: Division of Medical Oncology and Hematology,
`Princess Margaret Hospital, University Health Network, University of
`Toronto, Toronto, ON, Canada (CLT, LLS); Department of Biostatistics, The
`University of Texas M. D. Anderson Cancer Center, Houston, TX (JJL ) .
` Correspondence to: Lillian L. Siu, MD, FRCPC, Division of Medical Oncology
`and Hematology, Princess Margaret Hospital, University Health Network,
`University of Toronto, 610 University Ave, Ste 5-718, Toronto, ON, Canada
`M5G 2M9 (e-mail: lillian.siu@uhn.on.ca )
` See “Funding” and “Notes” following “References.”
` DOI: 10.1093/jnci/djp079
` © 2009 The Author(s).
`This is an Open Access article distributed under the terms of the Creative Com -
`mons Attribution Non-Commercial License (http://creativecommons.org/licenses/
`by-nc/2.0/uk/), which permits unrestricted non-commercial use, distribution, and
`reproduction in any medium, provided the original work is properly cited.
`
`708 Review | JNCI
`
`Vol. 101, Issue 10 | May 20, 2009
`
`Coalition for Affordable Drugs IV LLC – Exhibit 1007
`
`

`
` Table 1 . Glossary of terms
`
` Term
`
` Cohort
` Starting dose
` Dose increment (decrement)
` Dose-limiting toxicity (DLT)
`
` Dose – efficacy curve
`
` Dose – toxicity curve
`
` Target toxicity level
`
` Maximum tolerated dose (MTD)
`
` Optimal biological dose (OBD)
`
` Recommended phase II dose
`
` Pharmacokinetics
`
` Pharmacodynamics
`
` Therapeutic index
`
`Definition
`
`Group of patients treated at a dose level.
`The dose chosen to treat the first cohort of patients in a phase I trial.
`The percent increase (or decrease) between dose levels.
`Toxic effects that are presumably related to the drugs that are considered unacceptable (because
` of their severity and/or irreversibility) and that limit further dose escalation. DLTs are defined before
` beginning the trial and are protocol specific. They are typically defined based on toxic effects seen in
`
`the first cycle and specified using a standardized grading criteria, for example, Common Terminology
` Criteria for Adverse Events.
`The dose – efficacy curve reflects the relationship between dose and probability of efficacy for an
` anticancer agent. A logistic function is commonly assumed to describe the dose – efficacy curve for
` cytotoxic agents and is characterized by a parameter, ␪ , which represents the slope of the
` dose – efficacy curve. Small values of ␪ indicate that the probability of efficacy increases very slowly
` with increasing dose levels, whereas large values of ␪ indicate a sharp increase in efficacy with
`
`increasing dose levels ( see Figure 1 ).
`The dose – toxicity curve reflects the relationship between dose and probability of toxicity for an
` anticancer agent. A logistic function is commonly assumed to describe the dose – toxicity curve for
` cytotoxic agents and is characterized by a parameter, ␪ , which represents the slope of the
` dose – toxicity curve. Small values of ␪ indicate that the probability of toxicity increases very slowly
` with increasing dose levels, whereas large values of ␪ indicate a sharp increase in toxicity with
`
`increasing dose levels ( see Figure 1 ).
`The maximum probability of DLT that is considered acceptable in the trial. The target toxicity level in
` phase I trials is typically between 20% and 33%.
`Phase I trials conducted in the United States: the highest dose level at which ≤ 33% of patients
` experience DLT.
` Phase I trials conducted in Europe and Japan: the lowest dose level at which ≥ 33% of patients
` experience DLT (a misnomer in the sense that the MTD is actually not a tolerable dose).
` Phase I trials that use model-based methods: the dose that produces the target toxicity level.
`Dose associated with a prespecified most desirable effect on a biomarker among all doses studied
`
`(eg, inhibition of a key target in tumor or surrogate tissue or achievement of a prespecified
`
`immunologic parameter).
`Phase I trials with a toxicity endpoint that are conducted in the United States: the MTD.
` Phase I trials with a toxicity endpoint that are conducted in Europe and Japan: one dose level below
`
`the MTD.
` Phase I trials in which the endpoint is a prespecified biological endpoint: the OBD.
`Pharmacologic effects of the body on the drug (ie, the time course of drug absorption, distribution,
` metabolism, and excretion).
`Pharmacologic effects of the drug on the body (eg, nadir neutrophil or platelet count, nonhematologic
`
`toxicity, molecular correlates, imaging endpoints).
`The dosage or range of dosages of a drug that is required to produce a given level of damage to critical
` normal tissues (toxicity) divided by the dosage or range of dosages that yields a defined level of
` antitumor effect (efficacy) ( see Figure 1 ).
`
`explore the reasons for this disconnect. Finally, we recommend
`ways to assign dose escalation methods to evaluate new drugs or
`drug combinations. These recommendations are based on pre-
`clinical information, existing knowledge of agents that target the
`same or similar molecular pathways, and the availability of
`resources to execute such methods.
`
` Rule-Based Designs
` The main characteristic of rule-based designs is that they do
`not stipulate any prior assumption of the dose – toxicity curve.
`These designs comprise the so-called “up-and-down” designs
`because they allow dose escalation and de-escalation. The first up-
`and-down design was introduced in the late 1940s by Dixon and
`Mood ( 6 ), and Storer ( 7 ) described implementation of this design
`in clinical practice half a century later. The general principle of
`this design is to escalate or de-escalate the dose with diminishing
`fractions of the preceding dose depending on the absence or
`
` presence of severe toxicity in the previous cohort of treated
`patients ( Figure 2, A ). The simple up-and-down design converges
`to a dose that corresponds to a probability of severe toxicity of
`approximately 50%, which is higher than the 33% threshold com-
`monly accepted in most phase I cancer clinical trials. Although
`variations of this up-and-down design have been developed in an
`attempt to increase patient safety and to use toxicity data collected
`in real time ( 8 , 9 ), these designs have not been used much in clinical
`practice because they risk exposing patients to unacceptable levels
`of toxicity. The first rule-based design to be used widely in clinical
`practice was the traditional 3+3 design. Variations of the tradi-
`tional 3+3 design that have been put into clinical use include the
`accelerated titration designs and the pharmacologically guided
`dose escalation (PGDE) method.
`
` Traditional 3+3 Design
` The traditional 3+3 design remains the prevailing method for con-
`ducting phase I cancer clinical trials ( 7 ). It requires no modeling of
`
`jnci.oxfordjournals.org
`
`JNCI
`
`| Review 709
`
`

`
`three, one of four, or two of fi ve patients, but the trial will termi-
`nate if three or more dose-limiting toxicities are observed.
` The main advantages of the traditional 3+3 design are that it is
`simple to implement and safe ( Table 2 ). In addition, the accrual of
`three patients per dose level provides additional information about
`pharmacokinetic interpatient variability. However, a disadvantage
`of this design is that it involves an excessive number of escalation
`steps, which results in a large proportion of patients who are
`treated at low (ie, potentially subtherapeutic) doses while few
`patients actually receive doses at or near the recommended dose
`for phase II trials. This latter point is illustrated in Table 3 , which
`presents the dose escalation method used as well as the number of
`dose levels in recent fi rst-in-human single-agent phase I trials for
`anticancer agents that were eventually (1992 – 2008) approved by
`the US Food and Drug Administration (FDA) for the treatment of
`solid tumors. Among 21 trials that used the traditional 3+3 design,
`more than half involved six or more dose levels.
`
` Accelerated Titration Designs
` Accelerated titration designs combine features from variations of
`the traditional 3+3 design and the model-based design. Because the
`patient assignment to doses is based on prespecified rules, we clas-
`sify accelerated titration designs as rule-based designs. Through
`simulations based on a stochastic model fit to data from 20 actual
`phase I trials of nine different drugs, Simon et al. ( 36 ) described
`one control design and three accelerated titration designs. The
`control design, design 1, is a standard 3+3 design with a 40% dose
`increment between successive cohorts of patients. Although the
`three accelerated titration designs, designs 2, 3, and 4, were created
`based on a statistical model as described ( 36 ), the assignment of
`patients to dose levels follows specific rules according to the
`observed toxicities at each dose level. Designs 2 and 3 allow 40%
`and 100% dose escalations, respectively, between single-patient
`cohorts until a dose-limiting toxicity or two moderate toxicities are
`observed during cycle 1, at which point dose escalation reverts to
`the more conservative one used in design 1. In design 4, the 100%
`dose escalation between single-patient cohorts in the accelerated
`phase reverts to design 1 when one dose-limiting toxicity or two
`moderate toxicities are observed during any cycle (not just during
`cycle 1). Intrapatient dose escalation is allowed during the acceler-
`ated phase of designs 2, 3, and 4 ( Figure 2, C ). In all three acceler-
`ated titration designs, the standard 3+3 design is used after the
`accelerated phase as a stopping rule, and then the described model
`is recommended to estimate the MTD with all toxicity data col-
`lected during the trial. In addition, the model recommended for
`use included a parameter for cumulative toxicity as well as a para-
`meter for interpatient variability, such that the accelerated titra-
`tion designs would provide information in these aspects. In
`practice, investigators often determine the MTD based on the
`conventional 3+3 escalation rule without fitting trial data to the
`model at the end of the trial. Consequently, the original model-
`based accelerated titration designs have been adapted primarily as
`rule-based designs in clinical practice.
` The accelerated phase in accelerated titration designs — in
`which only one patient is included per dose level — along with the
`possibility of intrapatient dose escalation theoretically reduce the
`number of patients who are treated at subtherapeutic doses
`
`Dose–efficacy curve
`Dose–toxicity curve
`
`1
`
`0.3
`
`0.1
`
`efficacy
`
`Probability of toxicity or
`
`0
`
`x
`
`1
`
`
`
` Figure 1 . Typical dose – toxicity and dose – effi cacy curves for cytotoxic
`agents. This example illustrates that at dose x , the probability of effi -
`cacy is 30% and the probability of toxicity is 10%; hence, the therapeutic
`index of the drug at dose x is 10% divided by 30% = 1/3.
`
`Dose
`
`the dose – toxicity curve beyond the classical assumption for cyto-
`toxic drugs that toxicity increases with dose. This rule-based
`design proceeds with cohorts of three patients; the first cohort is
`treated at a starting dose that is considered to be safe based on
`extrapolation from animal toxicological data, and the subsequent
`cohorts are treated at increasing dose levels that have been fixed in
`advance ( Figure 2, B ). Historically, dose escalation has followed a
`modified Fibonacci sequence in which the dose increments become
`smaller as the dose increases (eg, the dose first increases by 100%
`of the preceding dose, and thereafter by 67%, 50%, 40%, and
`30% – 35% of the preceding doses). In most cases, the prespecified
`dose levels do not fit the exact Fibonacci sequence as described in
`the 12th century ( 5 ). If none of the three patients in a cohort expe-
`riences a dose-limiting toxicity, another three patients will be
`treated at the next higher dose level. However, if one of the first
`three patients experiences a dose-limiting toxicity, three more
`patients will be treated at the same dose level. The dose escalation
`continues until at least two patients among a cohort of three to six
`patients experience dose-limiting toxicities (ie, ≥ 33% of patients
`with a dose-limiting toxicity at that dose level). The recommended
`dose for phase II trials is conventionally defined as the dose level
`just below this toxic dose level.
` Alternative rules besides “3+3” have been proposed, including
`the “2+4,” “3+3+3,” and “3+1+1” (also referred as “best of fi ve”)
`rules ( 10 ). In the “2+4” design, an additional cohort of four patients
`is added if one dose-limiting toxicity is observed in a fi rst cohort of
`two patients. The stopping rule is the same as in the traditional 3+3
`design. In the “3+3+3” design, a third cohort of three patients is
`added if two of six patients in the fi rst two cohorts experience a
`dose-limiting toxicity at a certain dose level. The trial terminates if
`at least three of nine patients experience a dose-limiting toxicity.
`The “best of fi ve” design is more aggressive than the traditional
`3+3 design in that one additional patient is added if one or even
`two dose-limiting toxicities are observed among the fi rst three
`patients. Another patient is added if two dose-limiting toxicities
`are observed among the four treated patients. Dose escalation is
`allowed if dose-limiting toxicities are observed among none of
`
`710 Review | JNCI
`
`Vol. 101, Issue 10 | May 20, 2009
`
`

`
`Dose
`
`A
`
`Dose
`
`B
`
`DLT
`
`1
`
`DLT
`
`DLT
`
`1
`
`1
`
`1
`
`SD
`
`1
`
`1
`
`1
`
`1
`
`RD
`
`DLT DLT
`
`3 3
`
`3
`
`3
`
`3
`
`RD
`
`3
`
`3
`
`3
`
`SD
`
`Time
`
`Time
`
`Dose
`
`C
`
`Dose
`
`D
`
`DLT
`
`DLT DLT
`
`3
`
`3
`
`1
`
`2
`
`3
`
`RD
`
`DLT DLT
`
`3 3
`
`3
`
`3
`
`RD
`
`3
`
`1
`
`Plasma drug AUC >
`prespecified treshold
`
`Determination of
`plasma drug AUC
`
`1
`
`1
`
`SD
`
`Intrapatient dose escalation
`
`1
`
`1
`
`SD
`
`Time
`
`Time
`
`Dose
`
`E
`
`Dose
`
`F
`
`DLT
`
`DLT
`
`1
`
`DLT
`
`DLT
`
`1
`
`1 1 1
`
`3
`
`RD
`
`1 1 1
`
`3
`
`RD
`
`1
`
`1
`
`Computation of p(DLT at next DL)
`= target toxicity level
`
`1
`
`SD
`
`1
`
`1
`
`1
`
`SD
`
`Computation of p(DLT at next DL)
`= target toxicity level
`Computation of p(DLT at next DL)
`= overdosing or excessive overdosing
`
`
` Figure 2 . Graphical depiction of dose escalation methods for phase I
`cancer clinical trials. Each box represents a cohort comprising the indi-
`cated number of patients treated at a given dose level. A ) Simple
`up-and-down design. B ) Traditional 3+3 design. C ) Accelerated titra-
`tion design. Dashed arrows represent intrapatient dose escalation.
` D ) Pharmacologically guided dose escalation. E ) Modifi ed continual
`
`Time
`
`Time
`
`
`reassessment method. F ) Escalation with overdose control. “Overdosing
`or excessive overdosing” refers to doses that exceed the MTD. DLT =
`dose-limiting toxicity; SD = starting dose; RD = recommended dose; DL
`= dose level; AUC = area under the curve for drug concentration as a
`function of time; p(DLT at next DL) = probability of dose-limiting toxicity
`at the next dose level.
`
`( Table 2 ). Permitting intrapatient dose escalation in accelerated
`titration designs is appealing because it gives some patients the
`opportunity to be treated at higher and presumably more effective
`doses. For example, in the fi rst-in-human phase I trial of ixabepi-
`lone, which used an accelerated titration design with intrapatient
`dose escalation, all patients received the drug at the eventually
`established recommended dose for phase II trials ( 35 ). On the
`other hand, unless the model recommended in the original publi-
`cation (36) using parameters for cumulative toxicity and interpa-
`tient variability is applied and fi ts the data well, one drawback of
`
`intrapatient dose escalation is that it may mask the cumulative
`effects of treatment or, at the very least, would make them harder
`to differentiate from chronic or delayed toxic effects. However,
`regardless of the trial design used, chronic, delayed, or cumulative
`toxic effects are generally not well captured by most phase I trials
`because most patients with advanced cancers do not remain on
`study for extended periods of time. Furthermore, it can be diffi cult
`to present and interpret results of trials that allow intrapatient dose
`escalations because a single patient may contribute data for several
`dose levels.
`
`jnci.oxfordjournals.org
`
`JNCI
`
`| Review 711
`
`

`
` Table 2 . Theoretical main advantages and drawbacks of dose escalation methods for phase I cancer clinical trials *
`
`Advantages
`
`Drawbacks
`
` Dose escalation method
`
` Rule-based designs
`
` Traditional 3+3 design
`
`
`Easy to implement and safe
`
` Provide some data on PK interpatient variability
`
`
`Many patients treated at subtherapeutic doses
` Slow dose escalation
`Uncertainty about the RP2D
` Only the result from the current dose is used
`
`for determining the dose of next cohort of
` patients. Information on other doses is ignored.
`If model fitting is not performed (as is often the
` case in clinical practice):
`
` Intrapatient dose escalation may mask
`
` cumulative or delayed toxicities
`
` Difficult interpretation of the results when
`
`
`intrapatient dose escalation is allowed
`
` Uncertainty about the RP2D
`Need to obtain real-time PK results
`Interpatient variability may hamper dose
` escalation
`
`Need to have a prior guess of the RP2D
`
`Computations after each patient or cohort of
` patients
`Need real-time biostatistical support for dose
` escalation decisions (may also be an advantage)
`
`
`
` Accelerated titration designs
`
`More rapid dose escalation
`
`
`
`
` Pharmacologically guided dose
` escalation
`
` Model-based designs
`
` Modified continual reassessment
`
` method, escalation with overdose
`
` control, time-to-event continual
`
`
`reassessment method, EffTox,
`
` TriCRM
`
` May expose a greater proportion of patients at
` higher doses
` Data from all patients, cumulative toxicity, and
`
`interpatient variability can be fit to a model to
` establish the RP2D
`More rapid dose escalation
` Provide some data on PK interpatient variability
`
`
`Target toxicity level is explicitly defined
` More rapid dose escalation
` Use all available information from all patients
` Estimate of the RP2D with a confidence interval
` Take into account late-onset toxicities (time-to-event
` continual reassessment method)
` Take into account both toxicity and efficacy
`
`(EffTox + TriCRM)
`
` * PK = pharmacokinetic; RP2D = recommended phase II dose; EffTox = efficacy and toxicity method; TriCRM = an adaptative continual reassessment method that
`considers three potential trial outcomes: no efficacy and no toxicity, efficacy only, and toxicity only .
`
` Pharmacologically Guided Dose Escalation
` The PGDE method is another variation of the traditional 3+3
`design that has not been widely used in clinical practice. This
`approach assumes that dose-limiting toxicities can be predicted
`by plasma drug concentrations and that animal models can
`accurately reflect this relationship in humans ( 37 ). The PGDE
`method has two stages. A prespecified plasma exposure defined
`by the area under the curve for drug concentration as a function
`of time (AUC) is extrapolated from preclinical data. Then,
`pharmacokinetic data are obtained for each patient in real time
`to determine the subsequent dose level. As long as the prespeci-
`fied plasma exposure is not reached, dose escalation proceeds
`with one patient per dose level and typically at 100% dose
`increments (stage 1, Figure 2, D ). When the target AUC is
`reached or if dose-limiting toxicities occur, dose escalation
`switches to the traditional 3+3 design with smaller (usually
`around 40%) dose increments (stage 2).
` The PGDE method has not been widely adopted due to prac-
`tical obstacles, including: 1) logistic diffi culties in obtaining real-
`time pharmacokinetic results, which are required to determine
`the safety of the subsequent dose escalation; 2) problems in
`extrapolating preclinical pharmacokinetic data to phase I studies
`with different treatment schedules; and 3) risk of exposing the
`next patient to a highly toxic dose if the AUC obtained in the
`preceding patient was atypically low due to interpatient variabil-
`ity in drug metabolism. In clinical practice, the PGDE method
`has reliably defi ned the recommended dose for phase II trials for
`some cytotoxic agents such as certain anthracyclines and platinum
`
`compounds but has been found to be inappropriate for other
`classes of cytotoxic agents such as the antifolates, which display a
`high interpatient pharmacokinetic heterogeneity ( 38 ).
`
` Other Rule-Based Designs
` Several other rule-based designs have been proposed, including
`the isotonic regression model ( 39 ), the biased coin design ( 9 )
`and its variations ( 40 , 41 ), and the “rolling six” design ( 42 ). The
`rolling six design was originally proposed as a way to shorten
`the timeline of pediatric phase I trials by reducing the number
`of times a study is suspended to accrual ( 42 ). This method
`allows accrual of two to six patients concurrently onto a dose
`level based on the numbers of patients who are currently
`enrolled and evaluable, who experience a dose-limiting toxicity
`and who remain at risk of developing a dose-limiting toxicity.
`Because pediatric trials are typically conducted only after com-
`pletion of adult phase I trials, this design is intended to shorten
`the study duration in situations in which there is prior informa-
`tion about the dose range to be evaluated.
` Ji et al. ( 43 ) developed a rule-based design in which subse-
`quent patients are assigned to doses according to the toxicity
`outcome at the current dose by calculating the toxicity probabil-
`ity interval under the beta-binomial model. The authors also
`developed a freely available macro in Microsoft Offi ce Excel
`software that can be downloaded to facilitate the study conduct.
`Simulations have shown that the performance of this dose-
`fi nding design is better than the traditional 3+3 design and com-
`parable to some model-based designs.
`
`712 Review | JNCI
`
`Vol. 101, Issue 10 | May 20, 2009
`
`

`
` Table 3 . Characteristics of first-in-human phase I clinical trials for recent anticancer agents that were eventually approved by the
`US FDA*
`
` Agent
`
` Molecular targeted agents
`
` Trastuzumab
`
` Imatinib
`
` Gefitinib
`
` Erlotinib
`
` Cetuximab
`
` Bevacizumab
`
` Sorafenib
`
` Sunitinib
`
` Panitumumab
`
` Lapatinib
`
` Temsirolimus
` Cytotoxic agents
`
` Paclitaxel
`
` Vinorelbine
`
` Docetaxel
`
` Gemcitabine
`
` Topotecan
`
` Irinotecan
`
` Capecitabine
`
` Liposomal doxorubicin
`
` Temozolomide
`
` Oxaliplatin
`
` Pemetrexed
`
` Trabectedin
`
` Albumin-bound paclitaxel
`
` Ixabepilone
`
`Class or mechanism
`of action
`
`Year of FDA
`approval
`
`Dose escalation
`method
`
`Reason for
`stopping trial
`
`No. of
`patients
`
`No. of
`dose levels Reference
`
`
`
`
`Mab
`TKI
`TKI
`TKI
`Mab
`Mab
`TKI
`TKI
`Mab
`TKI
`STKI
`
`Anti-tubulin
`Alkaloid agent
`Anti-tubulin
`Antimetabolite
`Anti – topoisomerase I
`Anti – topoisomerase I
`Antimetabolite
`Anti – topoisomerase II
`DNA alkylating
`DNA alkylating
`Antimetabolite
`Alkaloid agent
`Anti-tubulin
`Anti-tubulin
`
`1998
`2001
`2003
`2004
`2004
`2004
`2005
`2006
`2006
`2007
`2007
`
`1992
`1994
`1996
`1996
`1996
`1998
`1998
`1999
`1999
`2002
`2004
`2004
`2005
`2007
`
`
`
`PK
`Traditional
`PK
`Traditional
`Toxicity
`Traditional
`Toxicity
`Traditional
`PK
`Traditional
`Target inhibition
`Traditional
`Toxicity
`Traditional
`Toxicity
`Traditional
`PK
`Traditional
`NR
`NR
`Toxicity
`Modified CRM
`
`
`Toxicity
`Traditional
`Traditional + IPDE Toxicity
`ATD
`Toxicity
`Traditional + IPDE Toxicity
`Traditional
`Toxicity
`Traditional
`Toxicity
`Traditional
`Toxicity
`Traditional
`Toxicity
`Traditional + IPDE Toxicity
`Traditional + IPDE Toxicity
`Traditional
`Toxicity
`Traditional
`Toxicity
`Traditional
`Toxicity
`ATD
`Toxicity
`
`
`
`
`
`
`
`18
`83
`64
`40
`52
`25
`69
`28
`96
`81
`24
`
`34
`20
`39
`47
`28
`17
`34
`26
`51
`23
`38
`21
`19
`21
`
`4
`14
`8
`5
`6
`5
`>5
`6
`13
`NR
`10
`
`11
`7
`6
`12
`5
`4
`5
`4
`15
`9
`10
`4
`4
`4
`
`(11)
`(12)
`(13)
`(14)
`(15)
`(16)
`(17)
`(18)
`(19)
`(20)
`(21)
`
`(22)
`(23)
`(24)
`(25)
`(26)
`(27)
`(28)
`(29)
`(30)
`(31)
`(32)
`(33)
`(34)
`(35)
`
` * FDA = US Food and Drug Administration; Mab = monoclonal antibody; TKI = tyrosine kinase inhibitor; STKI = serine/threonine kinase inhibitor; NR = not reported;
`PK = pharmacokinetic data; CRM = continual reassessment method; IPDE = intrapatient dose escalation; ATD = accelerated titration design.
`
` Summary of Rule-Based Designs
` The main advantages of rule-based methods are that they are
`easy to implement and do not require special software. However,
`their performance (operating characteristics) is not guaranteed and
`they have some drawbacks. For example, these designs may be
`inefficient in establishing the dose that meets a specific target tox-
`icity level. In addition, the decision of dose allocation for future
`patients as well as the definition of the recommended dose for
`phase II trials rely on information from the current dose level and
`do not use all available information. As such, the recommended
`dose for phase II trials is then selected from the prespecified dose
`levels depending on which one best fits the definition of acceptable
`toxicity set a priori. However, although not ideal, the rule-based
`methods have been successful in establishing safe recommended
`doses for phase II trials during the past several decades for antican-
`cer agents that were eventually used worldwide in clinical practice
`( Table 3 ).
`
` Model-Based Designs
` An alternative dose escalation method for phase I clinical trials is
`to use statistical models that actively seek a dose level that pro-
`duces a prespecified probability of dose-limiting toxicity by using
`toxicity data from all enrolled patients to compute a more precise
`dose – toxicity curve. This method can be conveniently carried
`out using Bayesian models. Bayesian models require an initial
`
` estimation of ␪ (also called prior distribution of ␪ ; Table 1 ), which
`characterizes the shape of the dose – toxicity curve. The occurrence
`of toxicity (or not) in patients enrolled at each dose level provides
`additional information for the statistical model and results in an
`adjustment of ␪ (also called posterior distribution of ␪ ) according
`to Bayes ’ theorem. The posterior distribution is then evaluated to
`identify the dose closest to the target toxicity level, and this dose is
`used to treat future patients and to set the recommended dose for
`phase II trials. These model-based designs use all of the available
`data to model the dose – toxicity curve, and they provide a confi-
`dence interval for the recommended dose for phase II trials at the
`end of the trial.
`
` Continual Reassessment Method and Modifications
` The continual reassessment method was the first Bayesian model –
` based method proposed for adoption in phase I trial designs ( 44 ).
`The initial estimate of ␪ required for this method is generally elic-
`ited from experts who are familiar with the preclinical data or who
`have experience with similar drugs if any exist. Although this initial
`estimate may not be accurate, it provides guidance about dose
`escalation. In the original description of the continual reassess-
`ment meth

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