throbber
STATISTICS IN MEDICINE
`Statist. Med. 17, 1103—1120 (1998)
`
`CANCER PHASE I CLINICAL TRIALS: EFFICIENT DOSE
`ESCALATION WITH OVERDOSE CONTROL
`
`JAMES BABB1*, ANDRE¤ ROGATKO1 AND SHELEMYAHU ZACKS2
`
`1Fox Chase Cancer Center, Department of Biostatistics, 510 Township Line Road, Cheltenham, PA 19012, U.S.A.
`2Binghamton University, Department of Mathematical Sciences, State University of New York, Binghamton, NY 13901, U.S.A.
`
`SUMMARY
`
`We describe an adaptive dose escalation scheme for use in cancer phase I clinical trials. The method is
`fully adaptive, makes use of all the information available at the time of each dose assignment, and
`directly addresses the ethical need to control the probability of overdosing. It is designed to approach the
`maximum tolerated dose as fast as possible subject to the constraint that the predicted proportion of
`patients who receive an overdose does not exceed a speci(cid:222)ed value. We conducted simulations to compare
`the proposed method with four up-and-down designs, two stochastic approximation methods, and with
`a variant of the continual reassessment method. The results showed the proposed method e⁄ective as
`a means to control the frequency of overdosing. Relative to the continual reassessment method, our scheme
`overdosed a smaller proportion of patients, exhibited fewer toxicities and estimated the maximum tolerated
`dose with comparable accuracy. When compared to the non-parametric schemes, our method treated fewer
`patients at either subtherapeutic or severely toxic dose levels, treated more patients at optimal dose levels
`and estimated the maximum tolerated dose with smaller average bias and mean squared error. Hence, the
`proposed method is promising alternative to currently used cancer phase I clinical trial designs. ( 1998
`John Wiley & Sons, Ltd.
`
`1. INTRODUCTION
`
`The primary purpose of a phase I clinical trial is to determine the dose of a new drug or
`therapeutic agent for use in a subsequent phase II trial. A long-accepted assumption underlying
`cancer therapy is that toxicity is a prerequisite for optimal antitumour activity.1 Consequently,
`one must endure some degree of treatment related toxic reaction if patients are to have
`a reasonable chance of favourable response. Since higher doses are associated with both greater
`therapeutic bene(cid:222)ts and an increased probability of severe toxic reaction, a cytotoxic drug should
`be administered at the maximum dose that cancer patients can tolerate. Consequently, the goal of
`a cancer phase I trial is to determine the highest dose associated with an acceptable level of
`toxicity. More precisely, the goal is to estimate the maximum tolerated dose (MTD), de(cid:222)ned as
`the dose for which the probability of a medically unacceptable, dose-limiting toxicity (DLT) is
`
`* Correspondence to: James Babb, PhD, Fox Chase Cancer Center, Department of Biostatistics, 510 Township Line
`Road, Cheltenham, PA 19012, U.S.A. E-mail: babb@canape.fccc.edu
`
`CCC 0277—6715/98/101103—18$17.50
`( 1998 John Wiley & Sons, Ltd.
`
`Received January 1996
`Revised May 1997
`
`INTELGENX 1031
`
`

`
`1104
`
`J. BABB, A. ROGATKO AND S. ZACKS
`
`equal to a speci(cid:222)ed value h:
`
`ProbMDLTD Dose"MTDN"h.
`The value chosen for the target probability h depends on the nature of the DLT; we would set it
`relatively high when the DLT is a transient, correctable or non-fatal condition, and low when it is
`lethal or life threatening.
`Representing the (cid:222)rst application of a proposed drug to humans, the phase I trial constitutes
`one of the most important steps in the drug(cid:213)s development.2 Since initial experience with a new
`agent may unduly in(cid:223)uence its fate, a careful and thoughtful approach to the design of phase I
`trials is essential. Unfortunately, clinical research involving humans poses serious ethical prob-
`lems and clinical trials involving oncology patients and cytotoxic drugs have been among the
`most problematic of all.3 In contrast to other phase I trials, cancer phase I trials have a thera-
`peutic aim. Typically, participants in a cancer phase I trial are patients at advanced disease stages
`who consent to participate in the trial only as a last resort in seeking cure. Thus, from
`a therapeutic perspective, one should design cancer phase I trials to minimize both the number of
`patients treated at low, non-therapeutic doses as well as the number given severely toxic
`overdoses.
`In the next section we describe a dose escalation scheme that controls the probability a patient
`will receive an overdose. The scheme, referred to as EWOC (escalation with overdose control), is
`Bayesian-feasible of level 1!a as de(cid:222)ned by Eichhorn and Zacks.4 That is, EWOC selects a dose
`level for each patient so that the predicted probability the dose exceeds the MTD is less than or
`equal to a speci(cid:222)ed value a. Zacks et al.5 showed that among designs that are Bayesian-feasible of
`level 1!a, EWOC is optimal in the sense that it minimizes the predicted amount by which any
`given patient is underdosed. Thus, EWOC is designed to approach the MTD as rapidly as
`possible subject to the constraint that the predicted proportion of patients given an overdose is
`less than or equal to a.
`
`2. METHOD
`
`The key concept underlying EWOC is that one can select dose levels for use in a phase I trial so
`that the predicted proportion of patients who receive an overdose is equal to a speci(cid:222)ed value a,
`called the feasibility bound. This is accomplished by computing, at the time of each dose
`assignment, the posterior cumulative distribution function (CDF) of the MTD. For the kth dose
`assignment the posterior CDF of the MTD is the function n
`k given by
`k(c)"ProbMMTD)c D Dk
`n
`N
`where Dk denotes the data at the time of treatment for the kth patient and would include for each
`previously treated patient the dose administered, the highest level of toxicity observed and any
`
`relevant covariate measurements. nk(c) is the conditional probability that c is an overdose given
`the data currently available. Based on this, EWOC selects for the kth patient the dose level
`xk such that
`
`k(xk)"a.
`n
`That is, we select the dose for each patient so that the predicted probability it exceeds the MTD is
`equal to a.
`
`( 1998 John Wiley & Sons, Ltd.
`
`Statist. Med. 17, 1103—1120 (1998)
`
`INTELGENX 1031
`
`

`
`DOSE ESCALATION WITH OVERDOSE CONTROL
`
`1105
`
`In the next section we describe EWOC for the speci(cid:222)c case where toxicity is measured on
`a binomial scale (presence or absence of DLT), there are no known covariates and one plans to
`accrue a (cid:222)xed number n of patients to the trial. Extensions of EWOC to accommodate more
`informative response measures, covariate information and variable sample sizes are currently
`under investigation. In Section 2.2 we present an example illustrating the application of EWOC
`to a cancer phase I clinical trial involving 5-(cid:223)uorouracil.
`
`2.1. Dose escalation method
`Let X.*/ and X.!9 denote the minimum and maximum dose levels available for use in the trial.
`One chooses these dose levels in the belief that X.*/ is safe when administered to humans and
`)MTD)X.!9.
`X.*/
`(1)
`The dose for the (cid:222)rst patient is X.*/ and we shall select only dose levels between X.*/ and X.!9 for
`use in the trial. Thus, if xi denotes the dose level selected for the ith patient, i"1, 2, n, then
`"X.*/
`x1
`3[X.*/, X.!9], "i"1, 2, n.
`xi
`We model the relationship between dose level and toxicity as
`#b
`ProbMDLTD Dose"xN"F(b
`1x)
`0
`where F is a speci(cid:222)ed distribution function, called a tolerance distribution, and b0 and b
`
`1 are
`’0 so that the probability of a DLT is a monotonic increasing
`unknown. We assume that b
`1
`function of dose. The MTD is the dose level, denoted c, such that the probability of a DLT is h. It
`follows from (2) that
`
`and
`
`(2)
`
`0
`
`1
`
`"X.*/
`
`1xi)yi[1!F(b
`We incorporate prior information about b0 and b
`
`
`
`F(b
`
`0
`
`c"F~1(h)!b
`b
`#F~1(h)!F~1(o
`0)
`b
`1
`0 denotes the probability of a DLT at the starting dose x1"X.*/. Figure 1 illustrates
`
`where o
`a typical dose-toxicity model.
`"0,Denote by yi the response of the ith patient where yi"1 if a DLT is manifest and yi
`
`
`
`otherwise. The data after observation of k patients is Dk"M(xi, yi), i"1, 2 , kN and the likeli-
`
`hood function of (b0, b
`1) given Dk is
`D Dk)" k<
`‚(b
`0, b1
`i/1
`
`
`
`1) de(cid:222)ned onh(b0, b
`)"M(a, b)3R2 : b’0, F(a#bX.*/))h)F(a#bX.!9)N.
`
`#b
`
`#b
`1xi)]1~yi.
`
`0
`
`1 through a prior probability density function
`
`( 1998 John Wiley & Sons, Ltd.
`
`Statist. Med. 17, 1103—1120 (1998)
`
`(3)
`
`INTELGENX 1031
`
`

`
`1106
`
`J. BABB, A. ROGATKO AND S. ZACKS
`
`Figure 1. The probability of DLT as a function of dose
`
`
`
`After an application of Bayes theorem, the joint posterior distribution of (b0, b1) given the data
`
`
`Dk is
`
`P(b
`
`0, b1
`
`
`
`where
`
`
`
`D Dk)"q~1‚(b0, b
`1
`
`D Dk)h(b
`
`
`
`0, b1)I)(b
`
`
`
`0, b1)
`
`(4)
`
`q"PP)
`‚(x, y D Dk)h(x, y)dx dy
`and I) denotes the indicator function for the set ). We can derive the marginal posterior
`cumulative distribution function of the MTD given Dk from (4) through the transformation
`„(b
`
`
`0, b1)"(o0, c). Denoting the image of ) under the transformation „ by „()), it follows from
`(1) and (3) that
`
`„())"[0, h]][X.*/, X.!9].
`
`The inverse transformation is
`
`„~1(o0, c)"( f1(o0, c), f2(o
`
`
`where the functions f1 and f2 are de(cid:222)ned on „()) by
`0)!X.*/F~1(h)
`
`f1(o0, c)"cF~1(o
`c!X.*/
`
`0, c))
`
`and
`
`0, c)"F~1(h)!F~1(of2(o
`0)
`
`c!X.*/
`We can now write the joint posterior probability density function (PDF) of (o0, c) given Dk as
`
`
`
`
`0, c D Dk)"q~1‚( f1(o0, c), f2(o0, c) D Dk)g(o0, c)
`P(o
`
`.
`
`( 1998 John Wiley & Sons, Ltd.
`
`Statist. Med. 17, 1103—1120 (1998)
`
`INTELGENX 1031
`
`

`
`DOSE ESCALATION WITH OVERDOSE CONTROL
`
`1107
`
`where
`
`D I„())(o
`F~1(p)K
`0, c)C L
`0, c)"h( f1(o0, c), f2(o0, c)) f2(o
`
`
`g(o
`0, c).
`Lp
`p/p0
`0, c) by the choice of h as the prior PDF of (b0, b1).
`
`Note that g is the prior PDF induced for (o
`
`Elicitation of prior information can be through speci(cid:222)cation of the PDF g directly, rather than
`through the choice of h. This might be advantageous since c is the parameter of interest and one
`often conducts preliminary studies at or near the starting dose so that one can select a meaningful
`informative prior for o
`0. Letting
`
`
`
`0.
`
`0, c)3„())N
`#(c)"Mo
`0 : (o
`we can write the marginal posterior PDF of the MTD given Dk as
`n(c D Dk)"PP#(c)
`P(o0, c D Dk) do
`The marginal posterior CDF of the MTD given Dk is then
`k(z)"P z
`n(c D Dk) dc, x3[X.*/, X.!9].
`
`n
`
`X.*/
`We can now describe EWOC as follows. The (cid:222)rst patient, or cohort of patients, receives the dose
`
`x1"X.*/. We select the dose for each subsequent patient so that on the basis of all the available
`data the posterior probability that it exceeds the MTD is equal to the feasibility bound a. Hence,
`the kth patient receives the dose
`
`K"2,2, n,
`
`(5)
`
`(6)
`
`"n~1
`m(k)(a)
`xk
`where m(k) denotes the number of observations available at the time of treatment for the kth
`patient.
`The dose sequence de(cid:222)ned by (5) assumes that all dose levels between X.*/ and X.!9 are
`available for use in the trial. However, due to practical and physical constraints, phase I clinical
`trials are typically based on a small number of prespeci(cid:222)ed dose levels. In such cases we select for
`the kth patient the dose level
`k(xk)!a)„
`!xk)„
`
`"maxMd1, 2, dr : di
`N
`1 and n
`Dk
`2
`where d1, 2, dr are the dose levels chosen for experimentation and „
`1 and „
`2 are prespeci(cid:222)ed
`non-negative real numbers we refer to as tolerances. We note that the dose sequence given by (6) is
`Bayesian-feasible of level 1!a if and only if at least one of the tolerances „
`
`1 and „2 is equal to
`zero. Positive tolerances would be chosen to permit the use of dose levels above yet suƒciently
`close to the optimal Bayesian-feasible dose xk.
`Since cancer patients often exhibit delayed response to treatment, the time required to resolve
`toxicity can be longer than the average time between successive accruals. Consequently, new
`patients frequently become available to the study before we have observed the responses of all
`previously treated patients. It is therefore important to note that EWOC does not require that we
`know all patient responses before we can treat a newly accrued patient. Instead, we can select the
`dose for the new patient on the basis of the data currently available.
`
`( 1998 John Wiley & Sons, Ltd.
`
`Statist. Med. 17, 1103—1120 (1998)
`
`INTELGENX 1031
`
`

`
`1108
`
`J. BABB, A. ROGATKO AND S. ZACKS
`
`Upon completion of the trial we can estimate the MTD by minimizing the posterior expected
`loss with respect to some choice of loss function l. Thus, the dose recommended for use in
`a subsequent phase II trial is the estimate cL
`l such that
`l(cL
`P X.!9
`l, c)n(cD Dn) dc)P X.!9
`l(x, c)n(cD Dn) dc "x3[X.*/, X.!9].
`
`X.*/
`
`X.*/
`
`Candidate estimators include the mean, median and mode of the marginal posterior PDF of the
`MTD. One should consider asymmetric loss functions since underestimation and overestimation
`have very di⁄erent consequences. Indeed, the dose xk selected by EWOC for the kth patient
`corresponds to the estimate of the MTD having minimal risk with respect to the asymmetric loss
`function
`
`if x)c that is, if x is an underdose
`la(x, c)"G a(c!x)
`if x’c that is, if x is an overdose.
`(1!a) (x!c)
`Note that the loss function la implies that for any d’0, the loss incurred by treating a patient at
`d units above the MTD is (1!a)/a times greater than the loss associated with treating the patient
`at d units below the MTD. This interpretation might provide a meaningful basis for the selection
`of the feasibility bound.
`We note that we can estimate the MTD using a di⁄erent prior PDF than that used to design
`the phase I trial.6 Furthermore, some authors (for example, Watson and Pelli7) have suggested
`use of Bayesian scheme to design the trial and use of maximum likelihood to estimate the MTD.
`
`(7)
`
`2.2. Example
`
`EWOC was used to design a phase I clinical trial that involved the antimetabolite 5-(cid:223)uorouracil
`(5-FU). In this trial a total of n"12 patients with malignant solid tumours are to be treated with
`a combination of 5-FU, leucovorin and topotecan. The goal is to determine the dose level of 5-FU
`that, when administered in combination with (cid:222)xed levels of the other two agents (20 mg/m2
`leucovorin, 0.5 mg/m2 topotecan), results in a probability of h"1/3 that a grade 4 hematologic
`or grade 3 or 4 non-hematologic toxicity is manifest within two weeks.
`Since preliminary studies indicated that 140 mg/m2 of 5-FU was well tolerated when given
`concurrently with up to 0)5 mg/m2 of topotecan, this level was selected as the starting dose for the
`phase I trial. Hence
`
`"140.
`X.*/
`Furthermore, a previous trial involving 5-FU alone estimated the MTD as 425 mg/m2. Since
`a given level of 5-FU has been observed to be more toxic when given in conjunction with
`topotecan than when administered alone, the MTD of 5-FU in combination with leucovorin and
`topotecan is believed to be below 425 mg/m2, the single agent MTD. Consequently, the max-
`imum allowed dose for the phase I trial is
`
`"425.
`X.!9
`Simulations and prior experience have shown EWOC to perform well when we take
`the tolerance distribution as the logistic and the prior PDF used for (o
`0, c) is the uniform
`distribution. Hence, we modelled the relationship between dose-limiting toxicity and the dose
`
`( 1998 John Wiley & Sons, Ltd.
`
`Statist. Med. 17, 1103—1120 (1998)
`
`INTELGENX 1031
`
`

`
`DOSE ESCALATION WITH OVERDOSE CONTROL
`
`1109
`
`level of 5-FU as
`
`#b
`1xN
`ProbMDLTD Dose"xN" expMb
`0
`#b
`1#expMb
`1xN
`0
`
`and we took the joint prior PDF of (o0, c) as
`
`0, c)"57~1I*0,0>2+]*140,425+(o
`0, c).
`g(o
`It follows that the marginal posterior CDF of the MTD at the time of the kth dose assignment is
`
`n
`
`k(x)"P x
`
`140
`
`P 0>2
`
`0
`
`
`
`P(o0, c D Dm(k)) do0 dc
`
`
`
`where
`
`with
`
`0, cD Dk)"q~1
`P(o
`
`
`expM f (o0, c D xi)N
`<
`i|S(k)
`m(k)<
`[1#expM f(o
`0, cD xi)N]
`i/1
`
`
`
`I*0,0>2+]*140,425+(o0, c)
`
`P <
`q"P
`T())
`logA o
`0
`1!o
`0, cD x)"
`f (o
`0
`
`i|S(k)
`
`m(k)<
`[1#expM f (o
`
`expM f (o0, c D xi)
`i/1
`
`B (c!x)#logA h1!hB (x!X.*/)
`c!X.*/
`
`
`
`0, cD xi)N]~1 do0 dc
`
`and
`
`"1N.
`
`S(k)"Mi"1, 2 , m(k) : yi
`For this trial the feasibility bound is equal to
`a"0)25
`this value being a compromise between the therapeutic aim of the trial and the need to avoid
`treatment attributable toxicity. Consequently, escalation of 5-FU between successive patients is
`to the estimated dose level that falls below the MTD with 75 per cent con(cid:222)dence. The (cid:222)rst patient
`"140 mg/m2. If the (cid:222)rst patient exhibits DLT, we will
`accrued to the trial receives the dose x1
`have violated the assumption that the initial dose is safe and we would suspend the trial (to restart
`at a lower initial dose or to terminate at the discretion of the principal investigator). Otherwise,
`we dose the next four patients according to the schedule given in Figure 2.
`
`3. SIMULATION COMPARISONS
`
`We conducted a simulation study to compare the performance of EWOC with seven phase I dose
`escalation schemes consisting of four up-and-down (UD) designs, two stochastic approximation
`(SA) methods and a Bayesian scheme known as the continual reassessment method (CRM). We
`compared the methods with respect to the proportion of patients assigned dose levels above and
`
`( 1998 John Wiley & Sons, Ltd.
`
`Statist. Med. 17, 1103—1120 (1998)
`
`INTELGENX 1031
`
`

`
`1110
`
`J. BABB, A. ROGATKO AND S. ZACKS
`
`Figure 2. The dose level to be given each of patients 2 to 5 in the 5-FU trial contingent on the doses and responses of
`preceding patients
`
`below the MTD, the proportion of patients exhibiting DLT and the bias and mean squared error
`observed for the estimator of the MTD.
`In cancer research the vast majority of phase I trials are based on UD dose escalation
`designs.8~11 With these methods, dose escalation proceeds either through a small number of
`preselected dose levels or via prespeci(cid:222)ed increments typically based on a modi(cid:222)ed Fibonacci
`sequence. The four designs considered in this study were proposed by Storer10 and are described
`as follows:
`”D1. Treat patients in cohort groups that consist of three patients each of whom receives the
`same dose. If no DLT is observed at a given dose level, then the next cohort receives the next
`highest dose. Otherwise, the next cohort is treated at the same dose and the trial then either
`continues at the next highest dose if exactly one of the last six patients exhibit DLT or
`terminates with observation of 2 or more DLT.
`”D2. Treat patients in cohorts of size three. Escalate the dose if no DLT is observed,
`de-escalate if more than one patient manifests DLT or repeat the dose if exactly one patient
`exhibits dose limiting toxicity. The trial continues until observation of a speci(cid:222)cied number n of
`patients.
`”D3. Treat patients one at a time. De-escalate the dose with each DLT and escalate it after
`two consecutive non-toxic responses. The trial continues until observation of a speci(cid:222)ed
`number n of patients.
`”D4. Treat patients one at a time. Escalate the dose continuously until observation of the (cid:222)rst
`DLT. Thereafter implement design UD3.
`
`( 1998 John Wiley & Sons, Ltd.
`
`Statist. Med. 17, 1103—1120 (1998)
`
`INTELGENX 1031
`
`

`
`DOSE ESCALATION WITH OVERDOSE CONTROL
`
`1111
`
`Various authors12~16 have proposed designs for phase I clinical trials based on the stochastic
`approximation methods of Robbins and Monro.17 With these designs patients are assigned doses
`according to
`xj‘1"xj
`
`!aj(yj
`P0 as jPR. The two SA
`N=
`where Maj
`j/1 is a sequence of positive real numbers such that aj
`methods considered in this simulation study correspond to the sequences
`
`!h)
`
`and
`
`where
`
`SA1: aj
`
`"1
`2j
`
`SA2: aj
`
`" 1
`jbj
`
`x2
`i
`
`j+
`
`(cid:236)(cid:239)(cid:239)(cid:239)(cid:237)(cid:239)(cid:239)(cid:239)(cid:238)
`
`2
`
`j+
`
`i/1
`
`"
`bj
`
`yiBNj"0
`
`!A j
`xiyi
`
`+
`i/1
`
`xiBA j
`
`+
`i/1
`
`j+
`
`i/1
`
`if
`
`otherwise.
`
`+
`i/1
`
`+
`i/1
`
`yiBNj
`xiBA j
`!A j
`xiyi
`!A j
`xiB2Nj
`
`+
`i/1
`i/1
`O(cid:213)Quigley et al.18 proposed a Bayesian dose escalation scheme referred to as the continual
`reassessment method (CRM). In its original formulation we describe CRM as follows. Specify
`a model for the dose—toxicity relationship and, at each stage of the trial, obtain a Bayesian
`estimate of the MTD using all the data then available. Choose the dose recommended for each
`patient from a prespeci(cid:222)ed discrete set of levels as close as possible to the estimate of the MTD.
`We simulated a modi(cid:222)cation of this scheme wherein we treat each patient at the mean of the
`marginal posterior PDF of the MTD. Note that the dose assigned to each patient by this scheme
`corresponds to the estimate of the MTD having minimal risk with respect to squared error loss,
`whereas the dose assigned by EWOC is chosen to minimize risk with respect to the asymmetric
`loss function given by (7).
`
`3.1. Simulation set-up
`
`Throughout the simulations we generated the data according to the logistic model
`
`D#lnCh(1!o
`expGlnC o
`0(1!h)D xcH
`
`0)
`0
`1!o
`PrMDLTD Dose"xN"
`0
`D#lnCh(1!o
`1#expGlnC o
`0(1!h)D xcH
`
`0)
`0
`1!o
`0
`with o
`0 assuming each of the values 0)05, 0)10 and 0)15, and the MTD taking on both of the values
`c"0)3 and c"0)5. Thus, we considered six cases corresponding to the six distinct combinations
`
`o
`
`o
`
`( 1998 John Wiley & Sons, Ltd.
`
`Statist. Med. 17, 1103—1120 (1998)
`
`INTELGENX 1031
`
`

`
`1112
`
`J. BABB, A. ROGATKO AND S. ZACKS
`
`0 and c. We assumed that dose levels were standardized so that the starting dose for each
`of o
`"0 and all subsequent dose levels were selected from the
`trial was the standardized dose x1
`unit interval. Since the performance of the UD designs depends on the dose levels available
`for use in the trial, we simulated each UD method using both of M0(0)2)1N and M0(0)1)1N
`as the set of preselected dose levels. Furthermore, since Storer10 constructed the UD designs
`for the speci(cid:222)c choice of target probability h"1/3, we used that value of h in all simulation
`runs.
`When simulating the Bayesian schemes, EWOC and CRM, we held the feasibility bound (cid:222)xed
`at a"0)25, we assumed that o
`0 and the MTD were independent a priori and we took the
`marginal prior PDF of the MTD as the uniform on the unit interval. To select a prior PDF for o
`0,
`we performed preliminary simulations to examine the performance of the two schemes when the
`prior for o
`0 was either
`
`P0 : the uniform on [0, h]
`
`or
`
`e3(0, h!o
`#e],
`0, o0
`Pe : the uniform on [o
`0).
`
`The results showed that for each selected value of e both CRM and EWOC exhibited more rapid
`dose escalation when we used Pe instead of P0 as the prior PDF. Furthermore, if for a given set of
`i!1 observations xi(e) denotes the dose recommended by EWOC for the ith patient when the
`(e
`
`prior PDF of o0 is Pe, then it can be shown that xi(e1)*xi(e
`2), "e1
`
`2. Consequently, for the
`
`class of priors considered here, the proportion of patients overdosed by EWOC is maximized if
`the ith patient is assigned the dose xi"lime?0xi(e), corresponding to the recommended dose
`
`under the assumption that o
`0 is known. Since the primary purpose of the simulation study was to
`compare the dose escalation schemes with respect to the frequency and magnitude of overdosing,
`we simulated both CRM and EWOC under the assumption that the probability of a dose-
`limiting toxicity at the starting dose was known. That is, to avoid conferring the model based
`schemes with an unfair advantage, we simulated EWOC and CRM under conditions unfavour-
`able to them.
`
`Upon completion of each UD, SA and CRM trial we estimated the MTD as cL "xn‘1, that is,
`
`as the dose level that we would have given the next patient had we allowed the trial to continue.
`This is the estimate typically used in practice. For the UD schemes it generally corresponds to the
`highest dose for which the observed proportion of DLTs was below h, while for CRM it has
`minimal risk with respect to squared error loss. A theoretical basis for the use of this estimate with
`the SA schemes is provided by the fact that xn converges almost surely to the MTD under rather
`general conditions.19 For CRM the estimate cL "xn‘1 is the marginal posterior mean of c given
`Dn. Hence, for the purpose of comparison, we used the posterior mean of c as the estimate of the
`MTD after each simulated EWOC trial. Furthermore, to determine whether the non-parametric
`schemes might compare more favourably to the Bayesian schemes if we had employed a model
`based estimate of the MTD, we used the posterior mean of c to obtain a second estimate of the
`MTD after each UD trial.
`0 and c, we simulated a total of 10,000 trials
`For each of the six combinations considered for o
`for each of the UD and SA designs while, due to their computational complexity, we simulated
`only 2000 trials for each of EWOC and CRM. Each simulated trial consisted of n"24 patients
`for each of the (cid:222)xed sample size methods (all but UD1) and a maximum of 30 patients for the
`variable length scheme UD1.
`
`( 1998 John Wiley & Sons, Ltd.
`
`Statist. Med. 17, 1103—1120 (1998)
`
`INTELGENX 1031
`
`

`
`DOSE ESCALATION WITH OVERDOSE CONTROL
`
`1113
`
`3.2. Results
`
`In Figures 3 to 12 we compare the dose escalation schemes on the basis of the proportion
`of patients treated at selected dose levels and with respect to the eƒciency with which they
`estimated the MTD. For Figures 3 to 5 and 7 to 12, each point represents the results observed
`0 and c, with the results obtained for c"0)3
`for one of the six selected combinations of o
`being depicted as a#and those for c"0)5 as a]. To facilitate comparison, we show the aver-
`age of the six results observed for each scheme (depicted as a f) and we have drawn a reference
`line through the average of the results obtained for EWOC. For each of the UD schemes we
`show the results obtained using the dose set M0(0)2)1N to the left of those obtained the dose set
`M0(0)1)1N.
`Figure 3 depicts the proportion of patients treated at dose levels for which the probability of
`a toxic response was less than 0)2. These patients were treated at dose levels where the drug is
`relatively inactive and therefore potentially non-therapeutic. The results indicate that EWOC
`treated fewer patients at low, possibly subtherapeutic, dose levels than did any of the non-
`
`parametric schemes. However, for each of the six selected combinations of o0 and c, we observed
`that EWOC treated more patients at the low dose levels than did CRM. This is expected since
`EWOC attempts to protect patients from being overdosed and so tends to treat patients at lower
`dose levels than does CRM.
`Figure 4 displays the proportion of patients treated at dose levels for which the probability of
`a toxic response was less than the target probability h but greater than 0)2. We describe these dose
`levels as optimal in the sense of being near but not above the MTD. The results show that the
`overall proportion of patients treated at optimal dose levels was higher for EWOC than it was for
`any of the other seven dose allocation schemes. Furthermore, for each of the six selected
`
`combinations of o0 and c, EWOC treated more patients at favourable dose levels than any of the
`other methods considered in this study except SA2.
`Figure 5 shows the proportion of patients treated at dose levels above the MTD. The overall
`proportion of patients overdosed by EWOC was 0)193, slightly below the selected feasibility
`bound a"0)25. For each of the six cases considered, EWOC overdosed a smaller proportion of
`patients than did CRM and compared favourably with SA1 and the UD schemes based on 6 dose
`levels. EWOC tended to overdose patients at a higher rate than SA2 and the UD schemes based
`on 11 dose levels. However, the latter schemes were overly conservative, treating a large
`proportion of patients at extremely low dose levels. Note that by choosing smaller values of a
`it is possible to reduce the proportion of patients overdosed by EWOC to levels that are
`comparable to those observed for any of the nonparametric designs. Figure 6 provides an
`indication of the extent to which EWOC a⁄orded protection from overdosing relative to CRM,
`"0)1 and c"0)3. There, we can see that EWOC treated only 31
`for the particular case where P1
`per cent of the patients at dose levels above the MTD, while CRM overdosed nearly twice as
`many. Relative to CRM, EWOC can have a substantial impact on the level of overdosing in
`a phase I clinical trial.
`Figure 7 shows the proportion of patients treated at dose levels for which the probability
`of a toxic response exceeded 0)5. These results show that EWOC subjected fewer patients to
`these severely toxic doses than did any of the other methods except SA2. When the MTD
`was equal to 0)5, CRM treated over three times as many patients at unacceptably high dose
`levels as EWOC. Overall, EWOC treated fewer patients at dose levels signi(cid:222)cantly above the
`target MTD.
`
`( 1998 John Wiley & Sons, Ltd.
`
`Statist. Med. 17, 1103—1120 (1998)
`
`INTELGENX 1031
`
`

`
`1114
`
`J. BABB, A. ROGATKO AND S. ZACKS
`
`Figure 3. Proportion of patients given doses for which the probability of a severe toxic reaction is less than or equal to
`1/5. Each]and#represents the results from all simulation runs for a particular parameter combination. Each point, (f),
`is the average of the results obtained for a particular method at the six parameter combinations considered. For each of
`the UD schemes the results obtained when only 6 dose levels were used are shown to the left of the results obtained when
`11 levels were used
`
`Figure 4. Proportion of patients given dose levels for which the probability of a severe toxic reaction is greater than 1/5
`but not greater than the target probability h"1/3 (see Figure 3 for symbol de(cid:222)nitions)
`
`Figure 5. Proportion of patients given dose levels above the MTD (see Figure 3 for symbol de(cid:222)nitions)
`
`( 1998 John Wiley & Sons, Ltd.
`
`Statist. Med. 17, 1103—1120 (1998)
`
`INTELGENX 1031
`
`

`
`DOSE ESCALATION WITH OVERDOSE CONTROL
`
`1115
`
`Figure 6. Histograms of the probability of toxic response for the dose levels selected by CRM and EWOC. For the
`intervals [0, 0)05), [0)05, 0)1), 2 , [0)9, 0)95), [0)95, 1)0] each histogram shows the proportion of patients given a dose level
`such that the probability of a severe toxic reaction is contained in that interval. The number in the body of each histogram
`in the proportion of patients given a dose level above the MTD
`
`( 1998 John Wiley & Sons, Ltd.
`
`Statist. Med. 17, 1103—1120 (1998)
`
`INTELGENX 1031
`
`

`
`1116
`
`J. BABB, A. ROGATKO AND S. ZACKS
`
`Figure 7. Proportion of patients given dose levels for which the probability of a severe toxic reaction was greater than 1/2
`(see Figure 3 for symbol de(cid:222)nitions)
`
`Figure 8. Average bias of c(cid:246) , the estimate of the MTD (see Figure 3 for symbol de(cid:222)nitions)
`
`Figures 8 to 11 compare the dose escalation schemes with respect to the eƒciency of c(cid:246) , the
`estimator of the MTD. The bias observed for each estimator (computed as cL !c) is shown in
`Figures 8 and 9 and the root mean squared error (JMSE) in Figures 10 and 11. In Figures 8 and
`10 the results are given for the non-parametric schemes when the MTD was estimated as the last
`recommended dose (that is, cL "xn‘1). Figures 9 and 11 compare the results obtained for EWOC
`and the UD schemes when the MTD was estimated as the mean of the marginal posterior
`distribution of c. All of the non-parametric methods tended to provide signi(cid:222)cantly biased
`estimates of the MTD while the overall mean bias associated with both of the Bayesian schemes
`was near zero. With respect to both bias and JMSE, EWOC provided more eƒcient estimates of
`the MTD than did any of the non-parametric methods. However, for each of the six cases
`considered, CRM estimated the MTD with smaller JMSE than did EWOC. This was anticip-
`ated, because by construction CRM should tend to select dose levels closer to the MTD, allowing
`more eƒcient estimation. Thus, a slight decrease in the accuracy of the MTD estimate is the price
`one pays for incorporation into EWOC of protection from overdosing.
`
`( 1998 John Wiley & Sons, Ltd.
`
`Statist. Med. 17, 1103—1120 (1998)
`
`INTELGENX 1031
`
`

`
`DOSE ESCALATION WITH OVERDOSE CONTROL
`
`1117
`
`Figure 9. Average bias of c(cid:246) , the estimate of the MTD (see Figure 3 for symbol de(cid:222)nitions)
`
`Figure 10.

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