11.09.2013 Views

Accelerated titration designs - Biometric Research Branch

Accelerated titration designs - Biometric Research Branch

Accelerated titration designs - Biometric Research Branch

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

4<br />

<strong>Accelerated</strong> <strong>titration</strong> <strong>designs</strong><br />

Janet Dancey<br />

Investigational Drug <strong>Branch</strong>, CTEp, DCTD, NCI, Bethesda,<br />

Maryland, USA<br />

Boris Freidlin and Larry Rubinstein<br />

<strong>Biometric</strong> <strong>Research</strong> <strong>Branch</strong>, DCTD, NCI, Bethesda, Maryland, USA<br />

4.1 Introduction<br />

A decade ago, investigators in oncology had a clear interest in modifications to the<br />

standard phase I design to make it more effcient, to treat fewer patients at nontoxic<br />

dose levels (which may be less effcacious) and to increase the precision of phase II<br />

dose recommendations. This was the conclusion of the 1996 Joint Meeting of the<br />

US National Cancer Institute and the European Organization for the <strong>Research</strong> and<br />

Treatment of Cancer (1,2). At approximately the same time, a review of the recent<br />

phase I oncology literature revealed that few investigators were makng use of the<br />

innovative phase I tral <strong>designs</strong> developed over the previous decade (3), which were<br />

meant to accomplish these very objectives.<br />

Approximately five years previous to this, Sheiner and coworkers published a series<br />

of papers in which they argued for the use of dose-response models in the analysis<br />

of phase I trals (4-). Standard practice in oncology trals, among other fields, was to<br />

analyze the dose-toxicity relationship only in terms of the population as a whole, and<br />

to analyze it separately for each dose. Rarely were attempts made to fit a dose-toxicity<br />

model to the phase I data that accounted for interpatient and intrapatient varabilty<br />

Statistical Methods/or Dose-Finding Experiments Edited by s. Chevret<br />

(Q 2006 John Wiley & Sons, Ltd<br />

91


92 ACCELERATED TITRATION DESIGNS<br />

separately, acc0!l0dated the possibility of cumulative toxicity and allowed for the<br />

constrction of dose-toxicity curves for the sensitive as well as the typical patients.<br />

In addition, Sheiner argued for the use of intrapatient dose escalation, to maximize<br />

the possibilty of individual patients receiving effcacious doses and to increase the<br />

accuracy of the analysis of the phase I data. This was not commonly practised in<br />

oncology phase I trals.<br />

In response to the above, Simon et at. (7) developed a family of 'accelerated <strong>titration</strong><br />

<strong>designs</strong>' and proposed the use of an accompanying dose-toxicity model, based<br />

on the work of Sheiner and coworkers (4, 5). The main distinguishing features of<br />

these <strong>designs</strong> are (a) a rapid initial escalation phase, (b) intrapatient dose escalation<br />

and (c) the ability to analyze tral results using a dose-toxicity model that incorporates<br />

parameters for intrapatient and interpatient varation in toxicity and cumulative toxicity.<br />

The distinguishing features of the model are its simplicity and the incorporation<br />

of separate varables for interpatient and intrapatient varability, as well as for the<br />

possibility of cumulative toxicity.<br />

4.2 Design<br />

Simon et at. (7) proposed a family of accelerated <strong>titration</strong> dose-escalation <strong>designs</strong>.<br />

In their formulation all <strong>designs</strong> use 40 % dose-escalation steps. The dose-escalation!<br />

de-escalation rules are based on definitions of dose-limiting toxicity (DLT) and of<br />

'moderate' toxicity. These definitions may be protocol specific. For example, Simon<br />

et at. used any grade 2 toxicity that was considered to be treatment related as moderate<br />

toxicity. For purposes of comparson, they designated the standard phase I design<br />

(with 40 % escalation steps in place of the standard modified Fibonacci escalation)<br />

as 'design 1'. They then introduced accelerated <strong>designs</strong> designated as 'design 2',<br />

'design 3' or 'design 4' .<br />

Design 1 dictates that patients are dose escalated in cohorts of three until DLT is<br />

observed. One instace of DLT leads to treatment of three additional patients at the<br />

current dose level (with escalation continuing if no additional DLT is observed). Two<br />

instances ofDLT, at a dose level, leads to a halt in dose escalation, with the prior dose<br />

level declared the MTD so long as six patients have been treated at that level, with<br />

one instance of DLT (de-escalation continues until such a dose level is determned).<br />

Design 2 stars with an accelerated phase that uses single-patient cohorts per dose<br />

leveL. When the first instance of first-course DLT is observed or the second instance<br />

of first-course moderate toxicity is observed, the cohort for the current dose level is<br />

expanded to thee patients and the tral reverts to use of design 1 for fuer cohorts.<br />

Design 3 is similar to design 2 except that double-dose steps are used during the<br />

accelerated phase. Two 40 % dose steps correspond to approximately a doubling of<br />

the actual dose. The accelerated phase ends, as with design 2, when the first instance of<br />

first-course DLT or the second instance of first-course moderate toxicity is observed.<br />

After that, design 1 is used for fuher patients.<br />

I<br />

l<br />

l<br />

I<br />

I',<br />

i.,<br />

I .<br />

!


-<br />

ACCELERATED TITRATION DESIGNS<br />

93<br />

Design 4 is similar to design 3, except for the criterion that is used for trggering<br />

the end of the accelerated phase. With <strong>designs</strong> 2 and 3, the accelerated phase ends with<br />

the first-course instance of DLT or second instance of first-course moderate toxicity.<br />

With design 4, the trgger is the first instance of any-course DLT or the second instance<br />

of any-course intermediate toxicity. In addition, when the first instace of moderate<br />

toxicity is observed, two additional patients must have been treated at that dose, or a<br />

higher dose (during any course), without experiencing moderate or worse toxicity, in<br />

order that the accelerated phase continues. This may require the treatment of one or<br />

two additional patients at that dose. Hence, design 4 may stop the accelerated phase<br />

earlier than design 3.<br />

Table 4.1 summarzes the characteristics of the four dose escalation <strong>designs</strong>.<br />

Table 4.1 Summar of the four dose-escalation <strong>designs</strong> and the two intrapatient<br />

dose-escalation options.<br />

Design Description<br />

1<br />

2<br />

3<br />

4<br />

Escalation<br />

A<br />

B<br />

Cohorts of 3 new patients per dose leveL. If i of 3 patients<br />

experiences DLT in first course, expand to 6 patients<br />

Cohorts of i patient per dose leveL. When first instance of<br />

first-course DLT is observed, or second instance of first-course<br />

grade 2 toxicity of any type, expand cohort for current dose<br />

level and revert to use of design 1 for all furher cohorts<br />

Same as design 2, except that double-dose steps are used<br />

during initial accelerated stage of trial (both for<br />

between-patient and within-patient escalation)<br />

Cohort of 1 new patient per dose level and double-dose steps<br />

are used durng the initial accelerated stage of the tral. When<br />

the first instance of DLT is observed at any course, or the<br />

second instance of any-course grade 2 toxicity of any type,<br />

expand cohort for current dose level and revert to use of design<br />

1 for all further cohorts. When the first instance of moderate<br />

toxicity is observed, two additional patients must have been<br />

treated at that dose, or a higher dose (during any course),<br />

without experiencing moderate or worse toxicity, in order that<br />

the accelerated phase continues<br />

Description<br />

No withn-patient dose escalation. De-escalate if grade 3 or<br />

worse toxicity at previous course<br />

Escalate if grade 0-1 toxicity at previous course. De-escalate<br />

if grade 3 or worse toxicity at previous course<br />

-,


94 ACCELERATED TITRATION DESIGNS<br />

4.2.1 Intrapatient dose-escalation<br />

In order to maximize each patient's chance to be treated at a potentially active dose,<br />

the accelerated <strong>titration</strong> design allows intrapatient dose-escalation for a patient who<br />

remains on study and has no evidence of toxicity at the current dose. Specifically,<br />

the dose for the next course is escalated if less than moderate toxicity was observed<br />

for the patient during the current course. If moderate toxicity occured, then the dose<br />

stays the same for the next course for that patient. If DLT occurred, then the patient<br />

generally goes off-study, but if not, then the dose is reduced. For design 2, single-dose<br />

steps are used for intrapatient dose changes. For <strong>designs</strong> 3 and 4, double-dose steps<br />

are used for intrapatient dose changes durng the accelerated stage, and single-dose<br />

steps subsequently.<br />

All four <strong>designs</strong> may be used with and without intrapatient dose-escalation. Simon<br />

et at. (7) compared the performance of the four <strong>designs</strong>, with and without intrapatient<br />

dose-escalation, in terms of toxicity, potential effcacy (reduction of treatment at doses<br />

below the MTD) and tral length. Table 4.1 also summarzes the two intrapatient doseescalation<br />

options.<br />

4.3 Evaluation of performance<br />

Simon et at. (7) fit the above model to data from 20 phase I trals (involving 9 distinct<br />

agents). Only thee ofthe trals showed any evidence of cumulative toxicity (a :: 0).<br />

The estimates of a for the other trals were zero or very close to zero. The trials vared<br />

substantially in the other parameters and thus provide a broad range of experience for<br />

evaluation of the accelerated <strong>titration</strong> <strong>designs</strong>.<br />

Simon et at. (7) evaluated the performance of the accelerated <strong>titration</strong> <strong>designs</strong> by<br />

simulating phase I data based on the 20 sets of parameters estimated from the 20 real<br />

trals that they studied. For each of the 20 sets of parameters, they generated data for<br />

1000 phase I trals and applied each of their <strong>designs</strong> to the simulated data. Figure 4.1<br />

shows the average number of patients per tral utilzed by each of the <strong>designs</strong>. For<br />

each design, the average is taken over the same 20000 simulated datasets generated<br />

from the sets of parameters derived from the 20 actual trals analyzed. Results for<br />

eight <strong>designs</strong> are shown. Designs 1 to 4 are as described above. The <strong>designs</strong> labeled<br />

with B utilize intrapatient dose-escalation if the toxicity in the previous course is less<br />

than intermediate. Designs labeled with A do not permt intrapatient dose-escalation.<br />

Design lA corresponds to the standard design, although it does not use Fibonacci<br />

dose steps. Design lB is the standard design augmented to permt intrapatient doseescalation.<br />

As can be seen in Figure 4.1, the average number of patients is much greater<br />

for the standard design lA or lB than for any of the accelerated <strong>titration</strong> <strong>designs</strong>. The<br />

average number of patients is somewhat less for <strong>designs</strong> 3 and 4 that use doubledose<br />

steps compared to design 2. Although the average differences are not great, the<br />

differences for individual trals can be; i.e. for a tral in which the starng dose is<br />

very low relative to the dose at which intermediate toxicity is expected, <strong>designs</strong> 2<br />

and 3 wil require substantially fewer patients.


ACCELERATED TITRATION DESIGNS<br />

40<br />

35<br />

lJ 30<br />

ñi "¡:<br />

leC\<br />

25<br />

20<br />

Q)<br />

~<br />

Q) 15<br />

~<br />

10<br />

5<br />

0<br />

Patients and Cohorts Distribution for<br />

8 Designs<br />

Total<br />

patients<br />

Cohorts<br />

c: Design 1 A<br />

. Design 1 8<br />

o Design 2A<br />

c: Design 28<br />

. Design 3A<br />

. Design 38<br />

D Design 4A<br />

. Design 48<br />

Figure 4.1 Average number of patients and number of cohorts for the eight <strong>designs</strong>.<br />

Figure 4.1 also shows the average number of patient cohorts utilized by each<br />

design. The average is lowest for <strong>designs</strong> 3 and 4, which use double-dose steps.<br />

Although the difference in average number of cohorts is not large, the difference in<br />

average time to complete the trals wil be much shorter for <strong>designs</strong> 2 to 4 if patients<br />

are not instantaneously available, since the accelerated phase of those <strong>designs</strong> requires<br />

only one patient per cohort.<br />

Figure 4.2 shows the average number of patients experiencing each level of toxicity<br />

as their worst toxicity durg their treatment on the tral. With the standard design, an<br />

average of 23 patients experience less than intermediate toxicity (labeled 'no toxicity'<br />

in the figure). These patients are underteated. For design 2B the average number of<br />

underteated patients is about eight and for <strong>designs</strong> 3B and 4B the number is less than<br />

five. Ths major reduction in the number of underteated patients is achieved with very<br />

small increases in the average number of patients experiencing DLT or unacceptable<br />

toxicity with the accelerated <strong>titration</strong> <strong>designs</strong>. Figure 4.3 shows the average percentage<br />

of patients experiencing each level of toxicity as their worst toxicity during their<br />

treatment on the tral.<br />

The accelerated <strong>titration</strong> <strong>designs</strong> without intrapatient dose-escalation, 2A, 3A and<br />

4A, performed quite well with regard to reduction in average number of patients and<br />

reduction of number of underteated patients. However, they do not provide patients<br />

accrued early in the tral a full opportnity to be treated at a therapeutic dose. They<br />

are also less effective in situations where interpatient varabilty in susceptibility to<br />

toxicity is large.<br />

These <strong>designs</strong> may be attactive, however, when there is concern about cumulative<br />

toxicity. It is worth noting in ths regard that analysis of the 20 phase I trals used<br />

95


96<br />

en +-<br />

C<br />

'"§ Q)<br />

a. -0.. Q)<br />

.0<br />

E::<br />

Z<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

Figure 4.2 Average number of<br />

the eight <strong>designs</strong>.<br />

en<br />

+-<br />

c<br />

0.7<br />

0.6<br />

0.5<br />

'"§ Q)<br />

a. 0.4<br />

- 0+-<br />

c<br />

Q)<br />

(. ..<br />

Q)<br />

a.<br />

0.3<br />

0.2<br />

0.1<br />

Toxicity Distribution for 8 Designs<br />

+- ;: +- ;:<br />

~<br />

..<br />

'õ 'õ a. ;: Q) +-<br />

'x 'x Cl (. 'õ<br />

0 0 (.<br />

+- +- ct 'x<br />

0<br />

0<br />

~ c +-<br />

Z :J<br />

:2<br />

ACCELERATED TITRATION DESIGNS<br />

c: Design 1 A<br />

.. Design 18<br />

o Design 2A<br />

D Design 28<br />

.. Design 3A<br />

.. Design 38<br />

o Design 4A<br />

II Design 48<br />

patients with worst toxicity at each toxicity level, for<br />

Toxicity Distribution for 8 Designs<br />

0 ;: ;:<br />

+-<br />

+-<br />

..<br />

'õ 'õ ~ a. ;: Q) +-<br />

'x 'x Cl (. 'õ<br />

0 0 (.<br />

+- +- ct 'x<br />

0<br />

0<br />

:Q c +-<br />

:J<br />

Z ~<br />

o Design 1 A<br />

.. Design 18<br />

o Design 2A<br />

o Design 28<br />

.. Design 3A<br />

.. Design 38<br />

o Design 4A<br />

II Design 48<br />

Figure 4.3 Average percentage of patients with worst toxicity at each toxicity level,<br />

for the eight <strong>designs</strong>.<br />

l . '-'."_.'''' ~.<br />

--


-<br />

ACCELERATED TITRATION DESIGNS 97<br />

18<br />

16<br />

14<br />

ëi 12<br />

;:<br />

j 10<br />

~ 8<br />

8 6<br />

4<br />

2<br />

o<br />

Comparison of Design 1 A vs Design 48<br />

LO CJ C' I" ,. LO 0) C' I" ,. LO CJ C' I"<br />

,. ,. C\ C\ C\ C' C' 'V 'V 'V LO LO<br />

Number of Patients<br />

- Design 1 A<br />

- Design 48<br />

Figure 4.4 Diagram of the comparative performances of design 1A and design 4B,<br />

in terms of patients required to reach each dose level and to define the MID.<br />

for evaluation of these <strong>designs</strong> revealed no evidence of il effects from intrapatient<br />

dose-escalation and lead the investigators to conclude that 'cumulative toxicity does<br />

not appear to be a valid reason to prohibit intra-patient dose escalation, as it occurs<br />

rarely' (1).<br />

To ilustràte furter the effciency of the accelerated <strong>designs</strong> in comparson with<br />

the standard, we give in Figure 4.4 a simulated comparson of the performance of design<br />

4B versus design lA for a paricular dose-toxicity modeL. The accelerated design<br />

completes the tral with less than half the number of patients required by the standard.<br />

More dramatically, due to the single-patient cohorts and two-step escalations,<br />

it requires only one patient for every six of the stadard design to escalate through<br />

the portion of the dose-toxicity curve where DLT is unlikely. Of course, if the initial<br />

dose of the tral is not defined so conservatively, the comparson is not so extreme.<br />

4.4 Model-based analysis<br />

By using a model for the statistical distrbution of toxicity, based on current and<br />

previous doses, a graded toxicity scale, based on the unobserved continuous varable<br />

associated with toxicity and multi course treatment results, the accelerated <strong>titration</strong><br />

<strong>designs</strong> allow for an effcient approach to analysis of phase I data. The model used in<br />

Simon et at. (7) was based on measurng the worst toxicity experience for each patient<br />

durng each course of treatment; i.e. the model does not consider separate toxicity for<br />

each organ system, but takes the maximum over all organ systems' and records that<br />

worst toxicity separately for each course of treatment for each patient. The model<br />

was designed to represent different levels of worst toxicity. The toxicity experienced<br />

in a paricular course was determned by the curent dose administered and the total<br />

.--<br />

H<br />

r::<br />

j',i~:<br />

l-i'<br />

r¡:<br />

~;~l


98<br />

ACCELERATED TITRATION DESIGNS<br />

Table 4.2 Summar of the dose-toxicity model<br />

for both the unobserved continuous toxicity<br />

varable and the observed toxicity grade leveL.<br />

Model relating toxicity to dose<br />

Yij = log(dij + aDi) + ßi + êij<br />

dij = dose for the ith patient in course j<br />

a = cumulative toxicity parameter<br />

Dij = cumulative dose up to course j<br />

ßi = interpatient random effect, N (0, aÐ<br />

êij = intrapatient random effect, N(Q, a;)<br />

Yij -: K 1 grade 0-1 toxicity<br />

Ki -: Yij -: K2 grade 2 toxicity<br />

K 2 -: Yij -: K 3 grade 3 toxicity<br />

Yij ~ K3 grade 4 toxicity<br />

dose administered in the previous courses. The model incorporated parameters for<br />

both intrapatient and interpatient varabilty, and for cumulative toxicity.<br />

Suppose that the ith patient receives dose dij during dose j and received a total<br />

dose Dij for courses prior to j. Let a represent the effect of cumulative toxicity<br />

(a = 0 indicates no effect of cumulative toxicity). The random varable ßi represents<br />

interpatient varability in the toxic effects; ßi is taken to be normally distrbuted<br />

with mean zero and varance aJ. The random varable Bij represents intrapatient<br />

varabilty in the toxic response; B¡j is taen to be normally distrbuted with mean<br />

zero and varance a;. These terms and random varables determne the unobserved<br />

magnitude Yij of the worst toxicity for patient i in course j, according to the formula<br />

Yij = log(d¡j + aDij) + ßi + Bij.<br />

In addition to the thee parameters a, aJ and a;, there are also several parameters<br />

for converting value Yij into a graded level of toxicity. Values of Yij less than K 1<br />

correspond to less then moderate toxicity values between K 1 and K2 correspond to<br />

moderate toxicity, values between K2 and K3 correspond to dose-limiting toxicity and<br />

values greater than K3 correspond to life-theatening toxicity: If one does not wish<br />

to distinguish DLT from life-theatening toxicity, then only K 1 and K2 are needed.<br />

Therefore there are five or six parameters to be estimated from the data. Table 4.2<br />

summarzes the characteristics of the modeL. Ths model is a generalization of the K max<br />

model of Sheiner, Beal and Sambol (5) and of the model of Chou and Talalay (8,9).<br />

Given the data of the grade of toxicity (worst over organ systems) for each<br />

course of each patient, the method of maxmum likelihood is used to estimate<br />

the model parameters. Splus software for fitting the parameters is available at<br />

http://inus.nci.nih.govlbrb. That website also contains an Excel macro for managing<br />

. i.<br />

I<br />

l<br />

. j .<br />

. . ~-l<br />

.1<br />


~<br />

ACCELERATED TITRATION DESIGNS<br />

1<br />

0.9<br />

0.8<br />

~ 0.7<br />

:E 0.6<br />

B 0.5<br />

e 0.4<br />

D. 0.3<br />

Probabilties of Grade 3+ Toxicity<br />

(Chloroquinoxaline Sulfonamide)<br />

0.2<br />

0.1<br />

o OC\""COC:OC\,,<br />

.,.,.,.,.,C\C\C\<br />

Dose Level<br />

-Gr 3+ (16%tile)<br />

- Gr 3+ (50%tie)<br />

.. Gr 3+ (84%tie)<br />

Figure 4.5 Probabilities of grade 3+ toxicity at varous dose levels for the mean<br />

patient and the patients one standard deviation above and below the mean.<br />

dose assignments to patients durng accelerated <strong>titration</strong> design trals. The macro<br />

assists investigators in quality controlling the dose assignment and provides a convenient<br />

way of recording dose assignments in a systematic manner that makes the data<br />

available for subsequent analysis.<br />

Figure 4.5 ilustrates the power of the model-based analysis to constrct a dosetoxicity<br />

curve, not only for the typical patient (50th percentile) but also for the patient<br />

who falls one standard deviation below the typical in terms of increased susceptibility<br />

to toxicity (16th percentile). The standard approach to defining the MID is based on<br />

the probabilty of toxicity at a given dose for the population as a whole, which often<br />

roughly corresponds to the probabilty of toxicity for the typical patient. With this<br />

approach, the initial phase II dose would be set, for Figure 4.5, at dose level 16 or<br />

17, to keep the probabilty of DLT below 30 %. However, the model-based analysis<br />

reveals that such a dose level results in at least a 40-0 % likelihood of DLT for a<br />

nontrvial subgroup of the patient population (those at the 16th percentile or below).<br />

This might suggest that a more prudent approach would be to define a lower initial<br />

phase II dose, to accommodate the susceptibilty of this subgroup.<br />

Figure 4.6 ilustrates the power of the model-based analysis to constrct comparative<br />

dose-toxicity cures for the different levels of toxicity. For example, the<br />

analysis suggests that the dose-toxicity cures for grade 2 versus grade 3 toxicity are<br />

separated by approximately four dose levels. This indicates that for a given patient,<br />

as well as for the population as a whole, DLT is likely to occur approximately four<br />

dose levels beyond moderate toxicity, suggesting that accelerated dose-escalation is<br />

likely to be safe, both for the population and for a given individual. Even though the<br />

dose-toxicity curve for DLT is relatively steep, it is well separated from the curve for<br />

moderate toxicity.<br />

99


t.<br />

L-<br />

100<br />

1<br />

0.9<br />

0.8<br />

;: 0.7<br />

ŠO.6<br />

.g 0.5<br />

.c 20.4<br />

D. 0.3<br />

0.2<br />

0.1<br />

o<br />

ACCELERATED TITRATION DESIGNS<br />

Probabilties of Toxicity (50%tie Patient)<br />

(Chloroquinoxaline Sulfonamide)<br />

10 12 14 16 18 20 22 24<br />

Dose Level<br />

- Gr 2+ toxicity<br />

- Gr 3+ toxicity<br />

- Gr 4+ toxicity<br />

Figure 4.6 Probabilities of grade 2+, grade 3+ and grade 4+ toxicity at varous<br />

dose levels for the mean patient.<br />

Sheiner and coworkers (4,5) proposed the use of dose-toxicity models for phase I<br />

trals a decade ago. They are stil rarely used, despite their potential for facilitating<br />

the definition of a phase II staring dose.<br />

4.5 Clinical applications<br />

First-in-man phase I tral <strong>designs</strong> of oncology agents share the following characteristics:<br />

selection of a 'safe' starng dose, sequential dose-escalation in cohorts of<br />

patients and determnation of a recommended dose based on a prespecified primar<br />

endpoint, usually the occurrence of unacceptable toxicity in a defined number of patients<br />

treated at a given dose leveL. Optimal phase I <strong>designs</strong> result in the identification<br />

of a dose for furer evaluation in a manner that is both safe and effcient. Higher<br />

staring doses, fewer patients per dose level and large escalation steps require fewer<br />

patients overall. However, safety is enhanced by lower staing doses, more patients<br />

per dose level to ensure safety of the dose and smaller dosing increments. Phase I<br />

<strong>designs</strong> must strke a balance between these elements. <strong>Accelerated</strong> <strong>titration</strong> <strong>designs</strong><br />

proposed by Simon et aI. (7) use the following modifications to enhance effciency:<br />

as few as one patient per level are enrolled and initial dose-escalation steps are larger<br />

(e.g. 100 % increments in the absence of toxicity). The number of patients per dose<br />

level increases to thee, once toxicity of a minimum degree (e.g. second instace of<br />

grade 2; first instance of DLT) has been seen in at least one patient. Thereafter a<br />

minimum of thee patients in each cohort are recruited, expanding to six in the event<br />

that one of thee has a DLT in the protocol prescribed observation period (usually<br />

one cycle or 4-8 weeks of chronic therapy) and the dose-escalation increments are<br />

reduced.<br />

"";",:..,, ...~


~<br />

ACCELERATED TITRATION DESIGNS<br />

To assess the use and utilty of the accelerated <strong>titration</strong> <strong>designs</strong> in the evaluation<br />

of novel oncology therapeutics, we conducted a literature search using the ISI Web<br />

of Knowledge Database (Thomson ISI, Thomson Corporation, Philadelphia, Pennsylvania)<br />

in May and August 2005. All aricles in the database that cited the original<br />

paper by Simon et at. (7) were retreved and reviewed. In tota, 106 publications were<br />

identified. Aricles that focused on statistical methodology of phase I studies (10),<br />

were not of phase I studies (4), that evaluated combinations of agents (12), were<br />

review aricles (34), or of phase I studies that did not use the Simon et at. (7) accelerated<br />

<strong>titration</strong> <strong>designs</strong> (10), were not included in our review. In total, 36 publications<br />

of phase I trals of novel cancer therapeutics were identified. From the tral publications,<br />

the following details were abstracted: agent/class, schedule, type of design,<br />

study-specific modifications to the design (e.g. patients/cohort and dose-escalation<br />

increments during the accelerated phase, rules for termnating the accelerated phase,<br />

dose-escalation increments following termnation of the accelerated phase, dose levels<br />

evaluated and the number of dose levels evaluated during the accelerated phase<br />

and subsequently). A summar of these trals are provided in Tables 4.3 and 4.4.<br />

From the results of our review a number of observations can be made regarding the<br />

utilzation of the <strong>designs</strong>. Firstly, the classes of agents selected for evaluation using an<br />

accelerated <strong>titration</strong> design favor agents that belong to chemical classes that have been<br />

previously studied or to biological agents not associated with significant risk of severe,<br />

irreversible organ toxicity. This is not surprising, as agents with these characteristics<br />

would engender a level of comfort regarding the safety of using an accelerated <strong>titration</strong><br />

design. Secondly, <strong>designs</strong> 3 and 4, which utilize single-patient cohorts and 100 % doseescalations,<br />

are the most commonly used. Thirdly, almost half of the studies do not<br />

utilze intrapatient dose-escalation. Fourtly, the most common modifications to the<br />

<strong>designs</strong> are those determning the dose-escalation increments following termnation<br />

of the accelerated phase and/or modifications to rules for termnating the accelerated<br />

phase. Most trals with modifications in the dose increments following termnation<br />

of the accelerated phase stipulated dose increments of 15-30 % rather than 40 %.<br />

A few utilzed higher-dose increments (50-67 %) and a few reverted to a modified<br />

Fibonacci escalation schema. Rules for termnating the accelerated phase included<br />

the first occurrence of any toxicity, or the achievement of a prespecified dose (e.g.<br />

mouse equivalent MTD). Given the frequency and nature of these modifications,<br />

it appears that investigators retain concerns regarding the safety of the accelerated<br />

<strong>titration</strong> design dose-escalation increments and termnation rules.<br />

To assess the effciency and safety of the accelerated <strong>designs</strong>, the numbers of<br />

patients and dose levels, overall and during the accelerated phase, were evaluated<br />

across the 36 studies. Patients treated above the ultimately recommended phase II<br />

dose (RP2D) were identified. If a patient was treated at a dose level prescribed by<br />

the accelerated phase dose increase and that dose exceeded the RP2D, then the dose<br />

level was considered to have exceeded the recommended dose due to the accelerated<br />

phase rules. Similarly, patients that died on the study due to obvious or suspected<br />

treatment-related toxicity were identified. Those fatalities which occured at doses<br />

prescribed during the accelerated phase of the study were classified as deaths during<br />

101


f<br />

..<br />

0N Table 4.3 Phase I trials using accelerated <strong>titration</strong> design.<br />

Intra patient<br />

Author/ dose- Recommended<br />

reference Agent Schedule Design escalation dose<br />

LoRusso 5-Fluoro-pyrimidinone Orally daily for 5 days 4B Yes 625 mg/m2/day orally for<br />

eta!. (10) every 4 weeks 5 days every 4 weeks<br />

Goetz 17 -(Allylamino )-17- iv weekly x 3 every 2B Yes 308 mg/m2 weekly x 3<br />

et at. (11) demethoxygeldanamycin 4 weeks every 4 weeks<br />

Grem 17 -(Allylamino )-17- 1 hour is iv infusion 2B Yes 40 mg/m2 daily x 5 every<br />

et ai. (12) demethoxygeldanamycin daily for 5 days every 3 weeks<br />

3 weeks<br />

Sessa BBR3464, cationic iv daily x 5 every 4B Yes 0.12 mg/m2/day x 5 every<br />

et at. (13) triplatinum complex 3 weeks 3 weeks<br />

n<br />

Mross BBR3576, iv infusion every 4A No 150 mg/m2 every 4 weeks f)<br />

et at. (14) aza-anthrapyrazole 4 weeks<br />

Plummer BMS-184476, taxane analog iv weekly x 3 every 4B Yes 50 mg/m2/week x 2 every ~<br />

tr<br />

et at. (15) 4 weeks, later 21 days<br />

~<br />

amended to<br />

weekly x 2 every<br />

~<br />

""<br />

21 days<br />

""<br />

Mani BMS-247550, derivative of 1 hour iv infusion every Modified 4B Yes 40 mg/m2 every 3 weeks ~<br />

et at. (16) Epothilone B 3 weeks ~ ""<br />

Abraham BMS-247550, derivative of 1 hour iv infusion Modified 3B Yes 6 mg/m2/day x 5 every 0<br />

Z<br />

et ai. (17) Epothilone B daily x 5 every 21 days ei<br />

tr<br />

21 days<br />

cr<br />

""<br />

Gadgeel BMS-247550, derivative of 1 hour iv infusiwyery Modified 2B Yes 40 mg/m2 every 21 days ci<br />

Z<br />

et at. (18) Epothilone B 21 days<br />

cr<br />

.1:<br />

\~¿".~C':,.'~;<br />

,,;.:'.1<br />

.'<br />

,-~~\O¡.t~r''':'''~t'''''~~~~~t...~~~~....~~t~~ .


~<br />

Undevia CEP-2563, receptor 1 hour iv daily x 5 every Modified 3B Yes 256 mg/m2/day x 5 every<br />

et at. (19) tyrosine kinase inhibitor 21 days 21 days n<br />

Hovstadius CHS 828, cyanoguanidine orally once 4B Yes 20 mg once daily for tr<br />

r;<br />

et at. (20) daily x 5 days every 5 days (100 mg/cycle) ìO<br />

4 weeks every 4 weeks ~<br />

Rudek COL-3, matrix orally daily 4A No 36 mg/m2/d without m<br />

0<br />

et at. (21) metalloproteinase sunblock d<br />

inhibitor i-<br />

Syed COL-3, matrix Orally daily 2A No 50 mg/m2/day<br />

et at. (22) metalloproteinase ~ ¡.<br />

0<br />

inhibitor Z<br />

Rustin Combretastatin A4 iv x 5 for 2 weeks every Modified 4B Yes 52-68 mg/m2 x 5 for 0<br />

tr<br />

et at. (23) phosphate, ubulin 3 weeks 2 weeks every 3 weeks en<br />

¡.<br />

targeting agent a<br />

Z<br />

Chatterjee DRF-1042, camptothecin Orally daily x 5 for Modified 3B Yes 80 mg/m2 x 5 for 2 weeks en<br />

et at. (24) analog 2 weeks every every 3 weeks<br />

3 weeks<br />

Vilalona- Ecteinascidin-743, iv daily x 5 every Modified 3A No 325 g/m2/day daily x 5<br />

Calero tetrahydroisoquinoline 3 weeks<br />

et at. (25) alkaloid<br />

Ko et at. (26) EMD 273066 (huKS-IL2) iv daily x 3 every Modified 4A No 6.4 mg/m2/day x 3 every<br />

immunocytokine 4 weeks 4 weeks<br />

Goel GEM23 1 , oligonucleotide to 3 day (1 patient) or a Modified 4A No 120 mg/m2/day x 5 days<br />

et at. (27) type i regulatory subunit 5 day continuous iv<br />

of protein kinase A infusion<br />

Chen GEM23 1 , oligonucleotide to 2 hour iv infusions Modified 4A No 240 mg/m2 twice weekly<br />

et at. (28) type I regulatory subunit twice weekly<br />

of protein kinase A ..<br />

( Continued) 0 UJ<br />

i


~~.."" . -" .-~---~-~~.~_...<br />

0<br />

.-<br />

.t<br />

Table 4.3 ( Continued)<br />

Intra patient<br />

dose- Recommended<br />

Author/<br />

reference Agent Schedule Design escalation dose<br />

Borchmann H22xKi-4, bispecific IV days 1,3,5, 4B Yes 80 mg/m2 per cycle<br />

et al. (29) anti-CD30 and CD64 7-21 days<br />

monoclonal antibody<br />

0 ZC/<br />

C/ ..<br />

Gadgeel KRN5500, spicamycin 1 hour iV daily x 5 2B Yes 4.3 mg/m2/day x 5 every<br />

et al. (30) derivative every 3 weeks<br />

3 weeks<br />

Schoemaker MAG-CPT, polymer IV infusion over 3 days Modified 2A No 68 mg/m2/day for 3 days<br />

et at. (31) conjugate of every 4 weeks<br />

every 4 weeks<br />

camptothecin<br />

Matsumura MCC-465, doxorubicin 1 hour IV infusion every ATD No 32.5 mg/m2 every 3 weeks ~<br />

et al. (32) PEG immunoliposome 3 weeks planned n<br />

Clive Antagonist G substance P IV every 3 weeks until Modified 4A No 400 mg/m2 weekly 6 hour<br />

~<br />

et al. (33) analog the target maximum<br />

IV infusion<br />

plasma concentration<br />

~<br />

of 10 p,M then weekly<br />

~<br />

Matsumura NK9l1, micelle IV every 3 weeks Modified 3A No 50 mg/m2 every 3 weeks i- -i et at. (34) encaps!llated doxorubicin<br />

de Jonge PNU -159548, alky1cycline IV every 3 weeks Modified 4B Yes 12 and 14 mg/m2 IV every ~<br />

3 weeks in HP and MP -<br />

et al. (35)<br />

(2 studies)<br />

0<br />

patients<br />

Z<br />

ci<br />

Lockhar PNU -166196A, brostallcin, IV weekly x 3 every Modified 3B Yes 2.4 mg/m2/week tI<br />

et al. (36) a nonalkylating DNA 4 weeks<br />

minor groove binder<br />

';;.::,:,¡:<br />

\~;i::;\,~,.:" .<br />

~~#~,~ii~1¡¥.~'i'Niiml~.~~.,\'..i!~,.~~J!~~~,~'",~~~~,""".~'7'~~,"=-.,-,.~,.~...-,..,,.,./~.".,.....",.._...,,_.,,-.


t<br />

¡<br />

1 i<br />

I<br />

I<br />

r<br />

!<br />

i<br />

l<br />

i<br />

I l<br />

106<br />

ACCELERATED TITRATION DESIGNS<br />

Table 4.4 Summar of phase I studies using accelerated <strong>titration</strong> design.<br />

Trials (n)<br />

Patients (n)<br />

Design<br />

Intrapatient<br />

doseescalation<br />

Modifications<br />

Types of<br />

modifications<br />

Patients/study<br />

(n)<br />

Dose levels (n)<br />

Fold dose range<br />

Patients treated<br />

Above RP2D<br />

Dose levels<br />

Above RP2D<br />

Deaths due to<br />

toxicity<br />

Type<br />

1<br />

2<br />

3<br />

4<br />

Unknown<br />

Yes<br />

No<br />

Yes<br />

No<br />

Median<br />

5<br />

3<br />

o<br />

o<br />

Dose-escalation during<br />

the accelerated phase<br />

Patients/cohort during<br />

the accelerated phase<br />

Rules for termination<br />

of accelerated phase<br />

Dose-escalation<br />

following<br />

accelerated phase<br />

During accelerated phase<br />

o<br />

Range<br />

0-15<br />

0-12<br />

0-<br />

0-1<br />

(4 of 36 studies had a<br />

dose level that<br />

exceeded the RP2D<br />

durng the ATD)<br />

0-1<br />

(1 patient across all<br />

studies died due to<br />

toxicities)<br />

Median<br />

22<br />

6<br />

16<br />

6<br />

1<br />

For study<br />

o<br />

(13 patients across<br />

all studies died<br />

due to toxicities)<br />

36<br />

911<br />

Trials (n)<br />

1<br />

6<br />

10<br />

18<br />

1<br />

20<br />

16<br />

15<br />

11<br />

3<br />

2<br />

8<br />

19<br />

Range<br />

7-74<br />

3-15<br />

2-320<br />

0-28<br />

0-3<br />

0-3<br />

"<br />

,I'<br />

.,1i<br />

I<br />

1<br />

i<br />

" '.~<br />

;.-,'".-<br />

d


-<br />

ACCELERATED TITRATION DESIGNS 107<br />

the accelerated phase. As summarzed in Table 4.4, the accelerated <strong>titration</strong> <strong>designs</strong>,<br />

as used in these studies, rarely resulted in dose escalation beyond the recommended<br />

phase II dose. Only 4 of 36 exceeded the recommended phase II dose during the<br />

accelerated <strong>titration</strong> phase and only 1 death from toxicity occurred during the accelerated<br />

<strong>titration</strong> phase, among the 91 I patients enrolled in these studies. (It should be<br />

noted, however, that the use of acceleration may have contrbuted, in some trials, to<br />

exceeding the RP2D by a greater number of doses, or for a greater number of patients,<br />

than would otherwise have happened. Thus, the use of acceleration in these trials may<br />

have increased the overall number of patients treated above the RP2D, even beyond<br />

the acceleration phase itself, and thus contrbuted to a greater death rate from toxicity.)<br />

Based on its utilization in these selected studies, the accelerated <strong>titration</strong> design<br />

appears to provide an enhanced effciency with acceptable safety. However, there<br />

are a number of issues investigators might consider prior to selecting an accelerated<br />

<strong>titration</strong> design to evaluate a novel agent in a first-in-man phase I clinical tral.<br />

The use of minimum one-patient cohorts and larger dose-escalation steps may be<br />

advantageous under the following circumstances: (a) the agent is of a chemical class<br />

that has been widely studied; (b) tle agent is predicted to have minimal interpatient<br />

varability in pharacokinetics; (c) the agent's anticipated toxicity is unlikely to be<br />

severe or ireversible and is amenable to close monitoring and supportive interventions.<br />

Examples of agents most amenable to evaluation using a phase I accelerated<br />

<strong>titration</strong> design might be the following: a new formulation of a previously studied<br />

agent (e.g.liposomal formulation ofpaclitaxel), a biological agent with minimal toxicity<br />

based on animal models (e.g. antibody or small molecule inhibitor of a receptor<br />

tyrosine kinase inhbitor) or an agent for which significant interspecies varabilty in<br />

preclinical toxicology has led to a very conservative staring dose in a human phase I<br />

study. Under these circu:Qstances, the increased effciency and presumed safety of an<br />

accelerated design might make it preferable.<br />

Conversely, there are situations where an accelerated <strong>titration</strong> design may not<br />

provide the optimal balance between safety and effciency as either larger numbers<br />

of patients/dose cohort and/or smaller dose increments would be preferable. Agents<br />

associated with steep dose-response curves for toxicity, severe ireversible toxicity,<br />

unexplained mortalty in anmal toxicology studies or large varabilty in doses or<br />

plasma drg levels eliciting effects may require alternative <strong>designs</strong> to balance safety<br />

and effciency optimally. For example, larger patient numbers/dose cohorts may be<br />

preferred if there is anticipated wide interpatient varabilty in toxic effects due to<br />

pharacokinetic or pharacogenomic differences between patients. For this circumstance,<br />

larger patient numbers per dose level is appropriate since decisions about the<br />

safety of a given dose may require more than a single patient's experience. Similarly,<br />

when a pharacokinetic or a pharacodynamc endpoint, rather than toxicity, is the<br />

primar endpoint, larger numbers of patients per dose level are recommended due to<br />

anticipated interpatient varability in these endpoints. With either situation, the use<br />

of an accelerated <strong>titration</strong> design with single-patient cohorts may not be optimal.<br />

There are also situations where small dose-escalation increments may be advisable.<br />

For example, if the agent is predicted to have severe, ireversible or potentially


l~:r~<br />

:f!<br />

"".,"<br />

.~.;<br />

'0.1<br />

; ~ f<br />

"~~ ;<br />

~.~ ;<br />

:.;;.Î<br />

'31 !<br />

J.-r"<br />

~~ I<br />

~ ¡<br />

'/11<br />

108<br />

ACCELERATED TITRATION DESIGNS<br />

fatal organ toxicity based on animal toxicology, paricularly if associated with a steep<br />

dose-response curve for toxicity, relatively small changes in dose/concentration may<br />

lead from minimal toxicity to severe toxicity, and thus smaller dose-escalation increments<br />

are preferable to ensure safety.<br />

Approaches to enhancing the proportion of patients in a phase I tral receiving<br />

'therapeutic' dose levels includes not only limiting enrolment on lower dose levels but<br />

also allowing dose-escalation within individual patients. Intrapatient dose-escalation<br />

provides two advantages: it improves the lielihood of benefit from the agent for the,<br />

individual patient and it increases the experience at higher dose levels. <strong>Accelerated</strong><br />

<strong>titration</strong> <strong>designs</strong> proposed by Simon et at. (7) allow intrapatient dose escalation if no<br />

toxicity :: grade 1 was seen in the first cycle at the assigned dose leveL. Whle it did<br />

not appreciably shorten the study duration, it did allow more patients to be treated at<br />

or near the recommended phase II dose and increased the number of cycles evaluated<br />

at the higher dose levels.<br />

Although the rationale supportng intrapatient dose-escalation is appealing, it<br />

does not seem to be widely applied. Thus, despite the appeal of escalating patients<br />

to higher doses than they were assigned initially, should safety criteria be met, some<br />

issues remain with its routine application in phase I protocols. Many phase I protocols<br />

continue to be written prohibiting intrapatient escalation since it is believed<br />

to have a minimal impact on tral effciency while bringing with it concerns about<br />

practical issues regarding the 'rules' for implementing dose-escalation and safety.<br />

Studies that have allowed intrapatient escalation withn their protocols have generally<br />

allowed dose-escalation to occur after the patient has been evaluated at the current<br />

dose level for the duration of the observation period and where the patient has had<br />

minimal/no toxicity. Less commonly used are rules that require not only that the patient<br />

has not had significant toxicity but also that the next higher dose level has been<br />

evaluated in one or more new patients - a more strngent and cumbersome criterion<br />

that may be favored to enhance safety and also to distinguish between acute versus<br />

cumulative toxic events. Although experience with intrapatient dose-escalation<br />

withn phase I studies is limited, to date its use within phase I studies using an accelerated<br />

<strong>titration</strong> design did not appear to compromise patient safety or complicate<br />

the interpretation of the study results. Of note, withn a given protocol, it is important<br />

to require a minimum number of newly recruited patients at each dose level<br />

and to base furter dose-escalation decisions upon the behavior of the drg in these<br />

individuals.<br />

4.6 Conclusions<br />

<strong>Accelerated</strong> <strong>titration</strong> <strong>designs</strong> can dramatically reduce the number of patients accrued<br />

to a phase I tral, in comparson to the standard phase I design. They can also substantially<br />

shorten the duration of the phase I tral. With intrapatient dose-escalation<br />

and application of a dose-toxicity model, they provide much greater information than<br />

the stadard design and analysis with regard to cumulative toxicity, interpatient and<br />

~<br />

t...<br />

1


--<br />

ACCELERATED TITRATION DESIGNS 109<br />

intrapatient varability, steepness of the dose-toxicity curve and separation of the<br />

dose-toxicity curves for the varing toxicity levels. They also provide all patients<br />

entered in the tral a maximum opportunity to be treated at a therapeutic dose.<br />

Despite this, we find that the <strong>designs</strong> are not widely used, which is likely to<br />

be due to the conservativeness of investigators. Even when they are used, they<br />

are often used with an initial dose set much more conservatively than would be<br />

done for the standard design and without use of intrapatient dose-escalation, thereby<br />

reducing their effectiveness. A recent comprehensive review of the risk-benefit rela-<br />

tionship for phase I trals conducted over the past decade reveals an overall toxicity<br />

death rate of only O.OOS (46). An accompanying editorial (47) argues that<br />

such a low toxicity death rate, in the context of treatment for an often rapidly fatal<br />

disease, suggests that phase I trals may be conducted in an overly cautious<br />

fashion. Appropriate utilization of <strong>designs</strong> such as the accelerated <strong>titration</strong> <strong>designs</strong><br />

might increase the potential for benefit in phase I trals, with little increase in<br />

risk.<br />

References<br />

1. S.G. Arbuck (1996) Workshop on phase I study design. Ninth NCIÆORTC New Drug<br />

Development Symposium, Amsterdam, March 12, 1996. Ann. Oncology, 7(6), 567-<br />

73.<br />

2. E.A. Eisenhauer, P.J. O'Dwyer, M. Christian and J.S. Humphrey (2000). Phase I clinical<br />

tral design in cancer drug development. J. Clin. Oncology, 18(3),684-92.<br />

3. S.P. Dent and E.A Eisenhauer (1996) Phase I tral design: are new methodologies being<br />

put into practice? Ann. Oncology, 7(6), 561-6.<br />

4. L.B. Sheiner (1990) Implications of an alternative approach to dose-response trals. J.<br />

Acquired Immune Deficiency Syndrome, 3(Suppl. 2), 20-.<br />

5. L.B. Sheiner, S.L. Beal and N.e. Sambol (1989) Study <strong>designs</strong> for dose-ranging. Clin.<br />

Pharmacology Theory, 46(1), 63-77.<br />

6. L.B. Sheiner, Y. Hashimoto and S.L. Beal (1991) A simulation study comparng <strong>designs</strong><br />

for dose ranging. Statistics in Medicine, 10(3), 303-21 March.<br />

7. R. Simon, B. Freidlin, L. Rubinstein, S.G. Arbuck, J. Collns and M.e. Chrstian (1997)<br />

<strong>Accelerated</strong> <strong>titration</strong> <strong>designs</strong> for phase I clinical trals in oncology. J. Natl Cancer Inst.,<br />

89(15),1138-47.<br />

8. T.e. Chou and P. Talalay (1981) Generalized equations for the analysis of inhibitions of<br />

Michaelis-Menten and higher-order kinetic systems with two or more mutually exclusive<br />

and nonexclusive inhibitors. Eur. J. Biochemistry, 115(1), 207-16.<br />

9. T.e. Chou and P. Talalay (1984) Quantitative analysis of dose-effect relationships: the<br />

combined effects of multiple drgs or enzyme inhbitors. Adv. Enzyme Regulation, 22,<br />

27-55.<br />

10. P.M. LoRusso, S. Prakash, A. Wozniak, L. Flaherty, M. Zalupski, A Shields, H.<br />

Sands, R. Parchment and B. Jasti (2002) Phase I clinical tral of 5-fluoropyrimidinone<br />

(5FP), an oral prodrg of 5-fluorouracil (5FU). Investigational New Drugs, 20(1), 63-<br />

71.<br />

11. M.P. Goetz, D. Toft, J. Reid, M. Ames, B. Stensgard, S. Safgren, AA. Adjei, J. Sloan, P.<br />

Atherton, V. Vasile, S. Salazaar, A Adjei, G. Croghan and e. Erlichman (2005) Phase I


~'Y<br />

110<br />

ACCELERATED TITRATION DESIGNS<br />

tral of 17 -allylamino-17 -demethoxygeldanamycin in patients with advanced cancer J.<br />

Clin. Oncology, 23(6), 1078-87.<br />

12. J.L. Grem, G. Momson, X.D. Guo, E. Agnew, C.H. Takmoto, R Thomas, E. Szabo,<br />

L. Grochow, E GroUman, J .M. Hamlton, L. Neckers and RH. Wilson (2005) Phase I<br />

and pharacologic study of 17-( allylamno )-17 -demethoxyge1danamycin in adult patients<br />

23(9), 1885-93.<br />

with solid tumors. J. Clin. Oncology,<br />

13. C. Sessa, G. Capri, L. Gianni, E Peccatori, G. Grassell, J. Bauer, M. Zucchett, L. Viganò,<br />

A Gatti, C. Minoia, P. Liati, S. Van den Bosch, A Bemareggi, G. Camboni and S.<br />

Marsoni (2000) Clinical and pharacological phase I study with accelerated <strong>titration</strong><br />

design of a daily times five schedule of BBR3464, a novel cationic trplatinum complex.<br />

Ann. Oncology, 11(8),977-83.<br />

14. K. Mross, M.E. Scheulen, T. Licht, C. Unger, H. RicWy, AC. Stem, K. Kutz, M.G.<br />

Camboni, P. Barbieri, E. Verdi, B. Vincenzi and A Bemareggi (2004) Phase I clinical<br />

and pharacokinetic study of BBR 3576, a novel aza-anthapyrazole, administered Lv.<br />

every 4 weeks in patients with advanced solid tumors: a phase I study group tral of<br />

the Central European Society of Anticancer-Drug <strong>Research</strong> (CESAR). Anticancer Drugs,<br />

15(1), 15-22.<br />

15. R Plummer, M. Ghielmini, P. Calvert, M. Voi, J. Renard, G. Gallant, E. Gupta, H. Calvert<br />

and C. Sessa (2002) Phase I and pharacokinetic study of the new taxane analog BMS-<br />

184476 given weekly in patients with advanced malignancies. Clin. Cancer Res., 8(9),<br />

2788-97.<br />

16. S. Mani, H. McDaid, A Hamlton, H. Hochster, M.B. Cohen, D. Khabelle, T. Griffn,<br />

D.E. Lebwohl, L. Liebes, E Muggia and S.B. Horwitz (2004) Phase I clinical and pharmacokinetic<br />

study of BMS-247550, a novel derivative of epothilone B, in solid tumors.<br />

Clin. Cancer Res., 10(4), 1289-98.<br />

17. J. Abraham, M. Agrawal, S. Bake, A Rutt, M. Edgerly, EM. Balis, B. Widemann, L.<br />

Davis, B. Damle, D. Sonnichsen, D. Lebwohl, S. Bates, H. Kotz and T. Fojo (2003) Phase<br />

I tral and pharacokinetic study of BMS-247550, an epothilone B analog, administered<br />

intravenously on a daily schedule for five days. J. Clin. Oncology, 21(9), 1866-73.<br />

18. S.M. Gadgeel, A. Wozniak, RR Boinpally, R. Wiegand, L.K. Heilbrun, V. Jain, R<br />

Parchment, D. Colevas, M.B. Cohen and P.M. LoRusso (2005) Phase I clinical trial of<br />

BMS-247550, a derivative of epothilone B, using accelerated <strong>titration</strong> 2B design. Clin.<br />

Cancer Res., 11(17), 6233-9.<br />

19. S.D. Undevia, N.J. Vogelzang, AM. Mauer, L. Janisch, S. Mani and M.J. Ratain (2004)<br />

Phase I clinical tral of CEP-2563 dihydrochloride, a receptor tyrosine kinase inhibitor, in<br />

patients with refractory solid tumors. Invest. New Drugs, 22(4),449-58.<br />

20. P. Hovstadius, R Larsson, E. Jonsson, T. Skov, AM. Kissmeyer, K. Krasilnikoff, J. Bergh,<br />

M.O. Karlsson, A Lönnebo and J. Ahlgren (2002) A phase I study of CHS 828 in patients<br />

with solid tumor malignancy. Clin. Cancer Res., 8(9), 2843-50. -<br />

21. M.A Rudek, W.D. Figg, V. Dyer, W. Dahut, M.L. Turner, S.M. Steinberg, DJ. Liewehr,<br />

D.R. KoWer, J.M. Pluda and E. Reed (2001) Phase I clinical trial of oral COL-3, a matrx<br />

metalloproteinase inhibitor, in patients with refractory metastatic cancer. J. Clin. Oncology,<br />

19(2), 584-92.<br />

22. S. Syed, C. Takmoto, M. Hidalgo, J. Rizzo, J.G. Kuhn, L.A Hammond, G. Schwar,<br />

A. To1cher, A. Patnaik, S.G. Eckhardt and E.K. Rowinsky (2004) A phase I and pharmacokinetic<br />

study of Col-3 (Metastat), an oral tetracycline derivative with potent matrx<br />

metalloproteinase and antitumor properties. Clin. Cancer Res., 10(19),6512-21.


ii<br />

ACCELERATED TITRATION DESIGNS 111<br />

23. G.J. Rustin, S.M. Galbraith, H. Anderson, M. Stratford, L.K. Folkes, L. Sena, L. Gumbrell<br />

and P.M. Price (2003) Phase I clinical tral of weekly combretastatin A4 phosphate: clinical<br />

and pharacokinetic results. J. Clin. Oncology, 21(15), 2815-22.<br />

24. A. Chatterjee, R Digumari, RN. Mamdi, K. Katneni, V.V. Upreti, A. Surath, M.L.<br />

Srinivas, S. Uppalapati, S. Jiwatani, S. Subramanam and N.R Srinivas (2004) Safety,<br />

tolerabilty, pharacokinetics, and pharacodynamics of an orally active novel camptothecIn<br />

analog, DRF-1042, in refractory cancer patients in a phase I dose escalation<br />

study. J. CUn. Pharmacology, 44(7), 723-36.<br />

25. M.A. Vilalona-Calero, S.G. Eckhardt, G. Weiss, M. Hidalgo, J.H. Beijnen, C. van<br />

Kesteren, H. Rosing, E. Campbell, M. Kraynak, L. Lopez-Lazaro, C. Guzman, D.D.<br />

Yon Hoff, J. Jimeno and E.K. Rowinsky (2002) A phase I and pharacokinetic study<br />

of ecteinascidin-743 on a daily x 5 schedule in patients with solid malignancies. Clin.<br />

Cancer Res., 8(1), 75-85.<br />

26. Y.J. Ko, G.J. Bubley, R Weber, C. Redfern, D.P. Gold, L. Finke, A. Kovar, T. Dahl and<br />

S.D. Gilies (2004) Safety, pharacokinetics, and biological pharacodynamics of the<br />

immunocytokine EMD 273066 (huKS-IL2): results of a phase I tral in patients with<br />

prostate cancer. 1. Immunotherapy, 27(3), 232-9.<br />

27. S. Goel, K. Desai, A. Bulgar, A. Fields, G. Goldberg, S. Agrawal, R Marin, M. Grndel<br />

and S. Mani (2003) A safety study of a mixed-backbone oligonucleotide (GEM231) targeting<br />

the type I regulatory subunit alpha of protein kinase A using a continuous infusion<br />

schedule in patients with refractory solid tumors. CUn. Cancer Res., 9(11), 4069-76.<br />

28. H.X. Chen, J.L. Marshall, E. Ness, RR Marin, B. Dvorchik, N. Rizvi, J. Marquis, M.<br />

McKinlay, W. Dahut and M.J. Hawkins (2000) A safety and pharacokinetic study of<br />

a mixed-backbone oligonucleotide (GEM231) targeting the type I protein kinase A by<br />

two-hour infusions in patients with refractory solid tumors. CUn. Cancer Res., 6(4), 1259-<br />

66.<br />

29. P. Borchmann, R Schnell, L Fuss, O. Manzke, T. Davis, L.D. Lewis, D. Behnke, C. Wickenhauser,<br />

P. Schiler, V. Diehl and A. Engert (2002) Phase I tral of the novel bispecific<br />

molecule H22 x Ki-4 in patients with refractory Hodgkin lymphoma. Blood, 100(9),<br />

3101-7.<br />

30. S.M. Gadgeel, R.R Boinpally, L.K. Heilbrun, A. Wozniak, V. Jain, B. Redman, M. Zalupski,<br />

R Wiegand, R Parchment and P.M. LoRusso (2003) A phase I clinical tral of spicamycin<br />

derivative KR5500 (NSC 650426) using a phase I accelerated <strong>titration</strong> '2B'<br />

design. Investigational New Drugs, 21(1), 63-74.<br />

31. N.E. Schoemaker, C. van Kesteren, H. Rosing, S. Jansen, M. Swar, J. Lieverst, D. Fraier,<br />

M. Breda, C. Pellzzoni, R Spinell, M. Grazia Porro, J.H. Beijnen, J.H. Schellens and<br />

W.W. ten Bokkel Huinink (2002) A phase I and pharacokinetic study of MAG-CPT, a<br />

water-soluble polymer conjugate of camptothecin. Br. 1. Cancer, 87(6),608-14.<br />

32. Y. Matsumura, M. Gotoh, K. Muro, Y. Yamada, K. Shirao, Y. Shimada, M. Okuwa,<br />

S. Matsumoto, Y. Miyata, H. Ohkura, K. Chin, S. Baba, T. Yamao, A. Kannam, Y.<br />

Takamatsu, K. Ito and K. Takahashi (2004) Phase I and pharacokinetic study of<br />

MCC-465, a doxorubicin (DXR) encapsulated in PEG iinunoliposome, in patients with<br />

metastatic stomach cancer. Ann. Oncology, 15(3),517-25.<br />

33. S. Clive, D.J. Webb, A. MacLellan, A. Young, B. Byrne, L. Robson, J.E Smyth and D.L<br />

Jodrell (2001) Forear blood flow and local responses to peptide vasodilators: a novel<br />

pharacodynamc measure in the phase I tral of antagonist G, a neuropeptide growth<br />

factor antagonist. CUn. Cancer Res., 7(10), 3071-8.


112<br />

ACCELERATED TITRATION DESIGNS<br />

34. Y. Matsumura, T. Hamaguchi, T. Ura, K Muro, Y. Yamada, Y. Shimada, K Shirao, T.<br />

Okusaka, H. Ueno, M. Ikeda and N. Watanabe (2004) Phase I clinical trial and 91(10), phara-<br />

cokinetic evaluation ofNK911, a micelle-encapsulated doxorubicin. Br. J. Cancer,<br />

1775-81.<br />

35. MJ. de Jonge, J. Verweij, A van der Gaast, O. Valota, O. Mora, AS. Planting, M.A<br />

Mantel, S.V. Bosch, MJ. Lechuga, F. Fiorentini, D. Hess and C. Sessa (2002) Phase I and<br />

pharacokinetic studies of PNU-159548, a novel alycycline, administered intravenously<br />

to patients with advanced solid tumours. Eur. J. Cancer, 38(18), 2407-15.<br />

36. AC. Lockhar, M. Howard, KR. Hande, BJ. Roth, J.D. Berlin, F. Vreeland, A Campbell,<br />

E. Fontana, F. Fiorentini, C. Fowst, V.A. Paty, O. Lanord and M.L. Rothenberg (2004)<br />

A phase I dose-escalation and pharacokinetic study of brostallicin (PNU-166196A),<br />

a novel DNA minor groove binder, in adult patients with advanced solid tumors. CUn.<br />

Cancer Res., 10(2),468-75.<br />

37. AJ. Ten Tije, 1. Verweij, A Spareboom, A Van Der Gaast, C. Fowst, F. Fiorentini, J.<br />

Tursi, A Antonellni, M. Mantel, C.M. Harman, G. Stoter, A.S. Planting and MJ. De<br />

Jonge (2003) Phase I and pharacokinetic study of brostallicin (PNU-166196), a new<br />

DNA minor-groove binder, administered intravenously every 3 weeks to adult patients<br />

with metastatic cancer. CUn. Cancer Res., 9(8), 2957-6.<br />

38. J. Dupont, B. Bienvenu, C. Aghajanian, S. Pezzull, P. Sabbatini, P. Vongphrachan, C.<br />

Chang, C. Perkell, K Ng, S. Passe, L. Breimer, J. Zhi, M. DeMaro, D. Spriggs and S.L.<br />

Soignet (2004) Phase I and pharacokinetic study of the novel oral cell-cycle inhibitor<br />

Ro 31-7453 in patients with advanced solid tumors. J. CUn. Oncology, 22(16), 3366-<br />

74.<br />

39. R. Salazar, D. Bissett, C. Twelves, L. Breimer, M. DeMaro, S. Campbell, J. Zhi, S. Ritland<br />

and J. Cassidy (2004) A phase I clinical and pharacokinetic study of Ro 31-7453 given<br />

as a 7- or 14-day oral twice daily schedule every 4 weeks in patients with solid tumors.<br />

Clin. Cancer Res., 10(13),4374-82.<br />

40. S. Wadler, D. Makower, C. Clairmont, P. Lambert, K Fehn and M. Sznol (2004) Phase<br />

I and pharacokinetic study of the ribonucleotide reductase inhbitor, 3-aminopyridine-<br />

2-carboxaldehyde thiosemicarbazone, administered by 96-hour intravenous continuous<br />

infusion. J. CUn. Oncology, 22(9), 1553-63.<br />

41. 1. Murren, M. Modiano, C. Clairmont, P. Lambert, N. Savaraj, T. Doyle and M. Sznol<br />

(2003) Phase I and pharacokinetic study of trapine, a potent ribonucleotide reductase<br />

inhibitor, administered daily for five days in patients with advanced solid tumors. CUn.<br />

Cancer Res., 9(11), 4092-100.<br />

42. P.H. Jones, R.D. Burnett, i. Fainar, P. Nadolny, P. Walker, Z. Yu, D. Tang-Liu, T.S.<br />

Ganesan, D.C. Talbot, AL. Hars and GJ. Rustin (2003) A phase 1 study of tazarotene<br />

in adults with advanced cancer. Br. J. Cancer, 89(5), 808-15.<br />

43. S.E. AI-Batran, A Atmaca, F. Bert, D. Jäger, C. Frisch, A. Neumann, J. Ort, A Knuth<br />

and E. Jäger (2004) Dose escalation study for defining the maximum tolerated dose of<br />

continuous oral trofosfamide in pretreated patients with metastatic lung cancer. Onkologie,<br />

27(6), 534-8.<br />

44. E.C. Dees, S.D. Baker, S. O'Reily, M.A Rudek, S.B. Davidson, C. Ayleswort, K Elza-<br />

Brown, M.A Carducci and R.C. Donehower (2004) A phase I and phanacokinetic study<br />

of short infusions ofUCN-Ol in patients with refractory solid tumors. Clin. Cancer Res.,<br />

11(2 Pt 1), 664-71.<br />

-


T<br />

¡<br />

¡<br />

¡<br />

i<br />

I<br />

t<br />

I<br />

¡<br />

l<br />

¡<br />

t<br />

i<br />

I<br />

t<br />

¡ r<br />

l<br />

I ,<br />

r<br />

~<br />

r<br />

l<br />

i<br />

i<br />

I r<br />

t r<br />

Fe<br />

l<br />

a<br />

I<br />

l<br />

i<br />

~<br />

~<br />

~<br />

,f<br />

iIL<br />

ACCELERATED TITRATION DESIGNS<br />

45. B.C. Goh, M.J. Ratain, D. Bertucci, R Smith, S. Mani, N.J. Vogelzang, R.L. Schilsky,<br />

M. Hutchison, M. Smith, S. Averbuch and E. Douglass (2001) Phase I study of ZD9331<br />

on short daily intravenous bolus infusion for 5 days every 3 weeks with fixed dosing<br />

recommendations. J. Clin. Oncology, 19(5), 1476-84.<br />

46. E. Horstmann, M.S. McCabe, L. Grochow, S. Yamamoto, L. Rubinstein, T. Budd, D.<br />

Shoemaker, E.J. Emanuel and C. Grady (2005) Risks and benefits of phase 1 oncology<br />

trals, 1991 through 2002. N. Engl. J. Medicine, 352(9),895-904.<br />

47. R Kurzrock and RS. Benjamin (2005) Risks and benefits of phase I oncology trals,<br />

revisited. N. Engl. 1. Medicine, 352(9), 930-2.<br />

113

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!