Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Aug 14.
Published in final edited form as: Clin Cancer Res. 2013 May 15;19(10):2646–2656. doi: 10.1158/1078-0432.CCR-12-2939

Modeling the relationship between progression-free survival and overall survival: The phase 2/3 trial

Mary W Redman 1, Bryan H Goldman 1, Michael LeBlanc 1, Anne Schott 2, Larry Baker 2
PMCID: PMC4131693  NIHMSID: NIHMS458947  PMID: 23669424

Abstract

The standard phase 2 trial design has changed dramatically over the last decade. Randomized phase 2 studies have essentially become the standard phase 2 design in oncology for a variety of reasons. The use of these designs is motivated by concerns regarding the use of historical data to determine if a new agent or regimen shows promise of activity. However, randomized phase 2 designs come with the cost of increased study duration and patient resources. Progression-free survival (PFS) is an important endpoint used in many phase 2 designs. In many clinical settings, changes in PFS with the introduction of a new treatment may represent true benefit in terms of the gold standard outcome, overall survival (OS). The phase 2/3 design has been proposed as an approach to shorten the time to discovery of an active regimen. In this a paper design considerations for a phase 2/3 trial are discussed and presented in terms of a model defining the relationship between OS and PFS. The design is also evaluated using 15 phase 3 trials completed in SWOG between 1990 and 2005. The model provides a framework to evaluate the validity and properties of a employing a phase 2/3 design. In the evaluation of SWOG trials, 3 of 4 positive studies would have also proceeded to the final analysis and 10 of 11 negative studies would have stopped at the phase 2 analysis, if a phase 2/3 design had been used. Through careful consideration and thorough evaluation of design properties, substantial gains could occur using this approach.

Keywords: Progression-free survival, phase 2, phase 3, phase 2/3

Introduction

The standard phase 2 trial design has changed dramatically over the last decade. Randomized phase 2 studies have essentially become the standard phase 2 design in oncology for a variety of reasons. The use of these designs is motivated by concerns regarding the use of historical data to determine if a new agent or regimen shows promise of activity. These concerns stem from the preponderance of failed phase 3 trials in cancer research. When systematic differences exist between the study population and the historical control population or in the assessment of outcomes between the two populations, then evaluations of efficacy may be subject to bias, resulting in false leads or missed opportunities. While overall survival (OS) generally remains the primary endpoint in most phase 3 trials, progression-free survival (PFS) instead of response rate is increasing used in the phase 2 setting. As noted by Zhang et al (1), PFS assessment can be subject to bias based on the assessment schedule, consistency of evaluators, and a number of other factors.

Randomized Phase 2 studies can demand significant time and patient resources to conduct. (2,3) They are typically two to four times larger than single arm trials, and relative to phase 3 studies, they generally require similar levels of review and include the same degree of effort to initiate and conduct. Therefore, a well-designed randomized phase 2 trial may require a significant effort and time to complete, delaying the time to a definitive answer.

To address the afore-mentioned efficiency concerns, numerous authors have promoted a combination of a randomized phase 2 and phase 3 studies. (411) In essence, a randomized phase 2/3 design is a randomized phase 3 design with a very early look which can only stop for futility, but not for early signs of efficacy. (6) This early interim analysis may use an alternative endpoint other than the primary endpoint for the phase 3 trial; for example PFS may be used for the first interim analysis, when the primary endpoint for the final analysis is OS. At this interim analysis the go/no go decision conforms closely to the positive/negative conclusion from a typical randomized phase 2 trial, and as such there is a higher probability of stopping early for futility than with the typical early interim analysis.

In general, the most clinically meaningful outcome continues to be OS. However, as discussed by Korn and Crowley (12) and Villaruz and Socinski (13), PFS is becoming an endpoint thought to have clinical merit in its own right. In this article we discuss design considerations for a phase 2/3 study using PFS as the primary endpoint for the phase 2 component and OS as the primary endpoint for the phase 3. This necessitates making certain assumptions about the relationship between PFS and OS. Design options and qualifications are evaluated using a motivating example. In addition, to explore the validity of the proposed model, PFS and OS data from published phase 3 studies involving an FDA approved agent, or thought to be practice changing, are used. The paper concludes with an evaluation of the properties when applied to phase 3 trials completed by SWOG. To anchor this discussion of phase 2/3 designs we propose the following as the basic foundation for designing a phase 2/3 trial.

Design Considerations for Phase 2/3 Trials

The standard randomized phase 2 design using PFS as the primary endpoint will target larger improvements than the follow-on phase 3 using OS, and employ type I error rates between 10–20%. (14) While such a design may be based on improvement in PFS, interpretation of trial results often take into consideration multiple pieces of information such as OS, response, and toxicity. In contrast, a phase 2/3 requires the specification of one rule. The phase 2 component of a phase 2/3 trial is a modification of the stand-alone phase 2 design, using similar effect sizes and error rates. The first interim analysis in a phase 2/3 trial is considered the phase 2 analysis, with the analysis time determined based on the number of events defined by the phase 2 parameters. Futility is established at this interim analysis if the alternative hypothesis is rejected at 1-power of the phase 2 or equivalently, if the study fails to reject the null hypothesis at the type I error rate of the phase 2. Therefore in the design of a phase 2/3 trial it is important to evaluate the proposed model and design choices as a unified study rather than simply combining phase 2 and 3 study designs.

Steps to design a phase 2/3 trial:

  1. Determine if the setting is appropriate

  2. Define the assumed relationship between the phase 2 and phase 3 endpoints

  3. Define the phase 2 and phase 3 study parameters: target effect measure and size, type I and type II error rates

  4. Evaluate phase 2 designs in terms of feasibility/timeliness of analysis and impact on phase 3 design properties

  5. Evaluate the properties of the phase 2/3 design and adjust design parameters if necessary

Methods

Each specific step to design a phase 2/3 study is discussed below. Step 1 is discussed in terms of key considerations. Step 2 is discussed by presenting a possible model to define the association between PFS and OS. The discussion of Step 3 uses this model to determine the study parameters. The discussion of this step is further given by example of where a phase 2/3 design might be appropriate. A literature review of phase 3 studies reporting on PFS and OS is used to evaluate the proposed model. Finally, to evaluate the performance of a phase 2 interim analysis on completed phase 3 studies, a “phase 2 analysis” is performed on phase 3 studies led by SWOG between 1990 and 2005 which included OS as the primary outcome.

Determine if the setting is appropriate

The first step, determination of appropriateness, is less based on statistics and trial design issues than on subject matter knowledge. Specifically, within in a disease setting and treatment type, there has to exist an intermediate outcome (such as PFS) for which it is expected that a treatment effect on this outcome represents an effect on the primary outcome (such as OS). It would seem that the most appropriate setting would be in more advanced disease with limited numbers of effective therapies available for post-progression treatment. Of note, one could design a phase 2/3 study using the same endpoint for the phase 2 component as for the overall trial; however there will not be the same reduction in time and patient savings relative to using an earlier endpoint for the phase 2 component. In terms of evaluating the feasibility of this approach, the event rate is likely quite important because the basis for using such a design is time savings. If the rates are low, then in order for the sample size to in the phase 2 range, the phase 2 analysis would require a temporary closure to accumulate enough events. In this case, it is important to consider how feasible it would be to conduct such a study, because such an interim analysis may be more informative than the standard interim analysis and could impact the integrity of the trial.(15,16)

Define the assumed relationship between the phase 2 and phase 3 endpoints

Given that continuation of a phase 2/3 design past the phase 2 interim analysis is based solely on a rule defined for the phase 2 endpoint, it should be the case that differences in the phase 2 outcome capture the effect of treatment on the phase 3 outcome. In the context of PFS and OS, this assumption would indicate that all or almost all of the treatment benefit occurs before progression, and most of the difference in OS is due to differences in progression. This assumption is consistent with the criteria put forward by Prentice (17) in specifying the necessary conditions for a surrogate endpoint. Specifically, a key component of the criteria is that the surrogate endpoint captures any relationship between the treatment and the primary endpoint.

In order to describe the relationship between PFS and OS, we use the model proposed by Goldman and colleagues (18). The model is a mixture of survival distributions specified in terms of the time to progression (TTP), time to death without progression (Spre), and time to death following progression (Spost). Figure 1 depicts this model with \lambda_{P}^{c}\left( t\right) ,\lambda_{Pre}^{c}\left( t\right),\lambda_{Post}^{c}\left( t\right) representing the hazard rates at time t for the survival distributions for TTP, SPre, and SPost, for the control treatment (c=0) and the experimental treatment (c=1). The observed values for PFS and OS are: PFS = min{TTP, Spre) and OS = SPre if a patient dies before progression and OS = TTP+SPost, if the patient progresses before death. It follows, that if TTP and SPre are exponentially distributed, then so is PFS with hazard rate of \lambda_{P}^{c}+\lambda_{Pre}^{c}. However, as was shown in (18), in this model OS is not exponentially distributed even when there are constant rates of transition between the states defined by progressive disease or death, so that hazard rates for survival are not constant over time. The hazard ratio (HR) for OS in this model is the HR averaged over the observation time period.

Figure 1.

Figure 1

Proposed model for the association between progression and death

As depicted in Figure 1, treatment has an indirect effect on survival time through its effect on time to progression and a direct effect on death. If the main effect of treatment is through an indirect effect on survival due to an effect on progression, then progression represents a surrogate, and λPre is zero, or close to zero. However, perhaps even with a small risk of death before progression, it could well be assumed that \lambda_{Pre}^{c} does not vary across treatment arms, and then the PFS HR is smaller than the TTP HR. Additionally, if \lambda_{Pre}^{c}\left( t\right) is a function of time, then the PFS HR is not constant over time.

Define the phase 2 and phase 3 study parameters

Design of the phase 2 and phase 3 components require specification of the effect measure and target effect size, and the type I and type II error rates. The phase 2 parameters will be defined in terms of the phase 3 design. In this setting, the type I and type II error for the phase 2 component are more accurately stated in terms of the phase 3 endpoint: that is, the phase 2 type I error occurs if the null hypothesis that there is no difference in PFS is rejected when there is in fact no difference in OS. Likewise a phase 2 type II error occurs if the conclusion is that there is no difference in PFS when in fact there is a difference in OS.

Since the phase 2 interim analysis is much more aggressive than standard interim analysis boundaries, this analysis has a greater impact on the study design properties. For example if both phase 2 and 3 designs specify 90% power, the adjusted power is 81%. (6) Therefore, depending on the power of the phase 2, the adjusted power of the phase 3 could be significantly decreased.

Determination of the effect measures and sizes for the two outcomes is more complicated. Quite often, while studies are designed to evaluate the HR between two treatment arms, clinically meaningful benefits are defined in terms of the difference in medians or percentage alive at a given time point. Under the model proposed, the multiplier that defines the relationship between the hazard ratio for PFS and OS is a time-varying function of the post-progression hazard rate and the hazard rates for PFS on both treatment arms. When the hazard rate for survival post-progression is larger, that is when time between progression and death is shorter, the multiplier is closer to one and the hazard ratios for PFS and OS are more similar. It follows that in this situation, the ratio of medians is smaller than the average HR.

Assuming PFS is a perfect surrogate for OS, the HR for OS (\Lambda _{OS}) at time t can be shown to be product of the HR for PFS (\Lambda _{PFS}) and a function of the survival functions for PFS and survival post progression as follows:

  • \Lambda _{OS} &=&\Lambda _{PFS}\ast c,\ c<1..

Here the multiplier is generally less than 1, implying that the ΛPFS is generally some factor larger than \Lambda _{OS}. However, given that the target in clinical terms is usually both a hazard ratio and a difference in medians, the approach we use here is to “attribute” the targeted absolute difference in median OS to PFS to determine clinically meaningful hazard ratios. This does ignore that OS does not have proportional hazards in this model.

This model, while likely also not correct, highlights that in design and analysis the true model is generally unknown, but for practicality and consistency across studies, standard analysis approaches such as use of the log-rank test statistic or Cox regression are generally used. It is most likely that the deviations from proportional hazards and constant hazards will result in a reduction in power, but by averaging over the times, a meaningful measure is given. Again, this emphasizes the importance of evaluating the design using the proposed analysis approach.

Evaluate of feasibility and design properties

To demonstrate the evaluation of feasibility of design properties, the motivating example employed here is treatment of extensive stage small cell lung cancer (E-SCLC). The standard of care for E-SCLC has essentially been a platinum-agent (cisplatin) in combination with etoposide for more than 20 years. (19) Median survival is around 9–10 months, and median progression-free survival is around 5 months. Moreover, to date, only topotecan has been approved by the Food and Drug Administration for the second line treatment of SCLC, and its efficacy is quite modest. This highlights a setting where there is a desperate need to improve care and to do so quickly.

Using this example in E-SCLC, candidate designs for the phase 2 component of a randomized phase 2/3 were determined and compared with a stand-alone phase 2 and phase 3. A design targeting a 3-month improvement in median survival would be considered clinically valuable. Therefore, these designs targeted a 33% improvement in median survival, from 9 months to 12 months. Appling the 3 month difference to PFS, this would be equivalent to a 60% improvement in median PFS, from 5 to 8 months. The sample sizes were determined based on a uniform accrual rate of 20 patients/month. Four follow-up scenarios were considered: no, 3, 6, and 9 months of follow-up. A stand-alone randomized phase 2 would likely employ 9 months of follow-up. Table 1 details the required events and sample size for varying levels of type I and type II error rates.

Table 1.

Phase 2 Trial Designs for SCLC

Type I,
type II
error
Events Sample Size (Analysis time)
No follow-up 3 months follow-up 6 months follow-up 9 months follow-up
15%,15% 78 198 (10 mths) 150 (11 mths) 124 (13 mths) 108 (15 mths)
15%,10% 97 218 (12 mths) 180 (12 mths) 150 (14 mths) 132 (16 mths)
15%, 5% 130 266 (14 mths) 218 (15 mths) 192 (16 mths) 172 (18 mths)
10%,10% 120 256 (13 mths) 206 (14 mths) 178 (15 mths) 160 (17 mths)
10%, 5% 155 294 (16 mths) 252 (16 mths) 220 (18 mths) 200 (20 mths)
5%, 5% 197 350 (18 mths) 298 (19 mths) 268 (20 mths) 248 (22 mths)

Table 1 demonstrates that a phase 2/3 design is quite feasible in this setting with a relatively short time to the phase 2 analysis, and modestly small sample sizes even for the setting with no follow-up. In fact, because of the rapid event rate, for all of the designs the phase 2 interim analysis with no temporary closure for additional follow-up occurs at the same time or before the designs with follow-up, including the stand-alone randomized phase 2 design.

As discussed above, design of the phase 3 component of a phase 2/3 requires the choice of either a study with lower adjusted power or an increase in the phase 3 power to recover the power loss from the phase 2 analysis. A stand-alone phase 3 design with 81% power would require 506 patients with an expected analysis time of 38 months, whereas a phase 2/3 with adjusted 81% power (90%*90%) would require 638 patients with an analysis time of 44 months. A stand-alone phase 3 design with 86% power would require 570 patients with an expected analysis time of 41 months, whereas a phase 2/3 with adjusted 86% power (95%*90%) would require 764 patients with an analysis time of 51 months. These designs use the design parameters stated above, a 1-sided 2.5% log-rank test for significance (assuming exponential survival) and 12 months of follow-up. Therefore, using a stand-alone phase 2 with 10% error rates (see Table 1), a stand-alone phase 2 followed by a phase 3 with 81% power would require 666 patients (160+506) and a total study period of 55 months, whereas the phase 2/3 would require 638 patients and total study period of 44 months. Similarly, a phase 3 with 86% power would require 730 patients (160+570) and a total study period of 58 months for the stand-alone studies and 764 patients and a total study period of 51 months for the phase 2/3. Therefore this demonstrates in the given setting, a phase 2/3 design would reduce the time to a definitive answer relative to the traditional phase 2 followed by phase 3 designs.

Evaluation of PFS and OS in literature review

In the literature review of phase 3 studies, 8 breast cancer (2027), 13 colorectal (2840), 11 kidney (4151), 4 leukemia (5255), 10 lung and mesothelioma (5665), 1 lymphoma (66), 6 myeloma (6772) and 2 prostate cancer (7374) publications were identified. To evaluate the relationship between PFS and OS, the absolute difference in median PFS and OS, the ratio of median PFS values and the PFS HR, and the ratio of median OS values and the OS HR were compared using linear regression. These data are presented in Figure 2. Panel A presents the association between the difference in median PFS and OS. The blue line represents equality between change in OS and PFS and the red line presents the regression of the difference in median OS on the difference in median PFS. The regression coefficient for PFS is 0.89 and is statistically significant at level < 0.0001. Panel B presents the association between the PFS HR and the OS HR. Again, the blue line represents equality and the red the regression line. While the magnitude of association was significantly less than the association with the absolute difference in medians, the PFS HR was statistically associated with the OS HR at level 0.04, with a regression coefficient of 0.28. Panels C and D present the comparisons of the ratio of medians to the HRs for PFS and OS. Both values were highly statistically significant at level <0.0001 with coefficients of 0.72 for the PFS comparison and 0.70 for the OS comparison, indicating the HRs for PFS and OS are reasonably well represented by the ratio of medians.

Figure 2.

Figure 2

Comparison of measures of PFS and OS in literature review

Panel A: A comparison of the absolute difference in median PFS with the absolute difference in median OS.

Panel B: A comparison of the HR for PFS with the HR for OS

Panel C: A comparison of the ratio of median PFS values with the HR for PFS

Panel D: A comparison of the ratio of median OS values with the HR for OS

The blue lines represent equality between the X and Y axis, the purple lines represent the fitted regression values.

Phase 2/3 Evaluation of SWOG trials

SWOG provided an opportunity to evaluate this approach on real data. Fifteen phase 3 trials which included OS as the primary endpoint and had data on either PFS or relapse-free survival run through SWOG between 1990 and 2008 were identified. (7588) Table 2 includes a description each of these trials, including the disease setting, treatments, and design properties. Of these trials four were positive and the remaining 11 studies failed to reject the null hypothesis.

Table 2.

SWOG Trial Description

Study Design Observed Data
N Years Control
Arm
Outcomes
Target
HROS
α Power Accrual Control
Arm
Outcomes
Stop
Time
Result
Disease Accr FUP Years
S879782 Early stage
cervical
240 5.5 4 5-yr OS:
60%
1.78 0.05 0.85 262 6.3 5-yr OS:
66%
Final Positive
(GYN) 5-yr
PFS:
56%
(1.5)^ 5-yr
PFS:
62%
S900881 stomach or
GI junction
550 5.5 2 Med OS:
30m
1.4 0.025 0.9 573 7 Med OS:
27m
Final Positive
(GI) Med
RFS:
20m
Med
RFS:
19m
S930887 Advanced
NSCLC
400 2.5 1 Med OS:
6m
1.33 0.05 0.88 415 1.5 Med OS:
6m
Final Positive
Med
PFS: 2m
Med
PFS: 2m
S931380 high risk
stage I/II
3000 3.75 5 5-yr OS:
93%
1.45* 0.05 0.9 3126 3.1 5-yr OS:
88%
Final Negative
(Breast) 5-yr
DFS:
86%
5-yr
PFS:
80%
S932175 Multiple
myeloma
500+ 4 3 Med OS:
3y
1.33 0.05 0.81 519 9.8 Med OS:
4.9y
Final Negative
(Mmyel) Med
PFS:
1.3y
Med
PFS:
1.8y
S941585 high risk
stage II and
stage III
colon
1500 6.5 3.5 Med OS:
9.3y
1.35 0.05 0.95 1004 5 Med OS:
10.9y
Interim Negative
(GI) Med
DFS:
8.5y
Med
DFS:
9.4y
S942079 Colorectal 700 3.5 1.5 Med OS:
15m
1.33 0.025 0.9 723 4.1 Med OS:
13m
Final Negative
(GI) Med
PFS: 6m
Med
PFS: 5m
S943886 Non-Hodkin 206+ 5 2 2-yr OS:
40%
1.5 0.05 0.82 194 10 2-yr OS:
74%
Final Negative
(Lymph) 2-yr
DFS:
30%
2-yr
DFS:
73%
S950976 Advanced NSCLC 400 1.33 1 Med OS:
8m
1.5 0.025 0.94 415 1.8 Med OS:
9m
Final Negative
(Lung) Med
PFS: 4m
Med
PFS: 4m
S991683 advanced
refractory
prostate
620 3.5 1 Med OS:
18m
1.33 0.025 0.8 716 3.3 Med OS:
16m
Final Positive
(GU) Med
PFS: 4m
Med
PFS: 3m
S0001 glioblastoma
and
gliosarcoma
375 5 2 Med OS:
12m
1.4 0.05 0.925 179 4.2 Med OS:
10m
Interim Negative
(Brain) Med
PFS: 5m
Med
PFS: 4m
S000388 Advanced
NSCLC
500 1.67 1 Med OS:
8m
1.375 0.025 0.88 376 2 Med OS:
9m
Interim Negative
(Lung) Med
PFS: 4m
Med
PFS: 4m
S002377 Stage III,
NSCLC
672+ 3.5 2.5 Med OS:
21m
1.33 0.025 0.89 250 4.2 Med OS:
32m
Interim Negative
(Lung) Med
PFS: 7m
Med
PFS:
10m
S012478 Extensive
stage 1st line
620 4 1 Med OS:
10m
1.33 0.025 0.9 656 4.3 Med OS:
9m
Final Negative
(Lung) SCLC Med
PFS: 5m
Med
PFS: 5m
S020584 advanced
pancreas
704 5 1 Med OS:
6m
1.33 0.0125 0.92 743 2.2 Med OS:
6m
Final Negative
(GI) Med
PFS: 3m
Med
PFS: 3m
*

S9313 had DFS as a primary endpoint with HR=1.3 and no interim futility testing. A trial with the same characteristics but OS as a primary endpoint would use HR=1.45.

^

S8797 was powered for HR=1.78, but interim futility testing was based on HR=1.5.

+

Based on number randomized

NSCLC = Non-small cell lung cancer

GI = gastrointestinal

17-29 Reference number – see bibliography

Of the 15 trials, 6 trials were identified as occurring in disease settings where there would likely be little gain in time and patient accrual using this approach, given the assumed progression-free survival times in the control arms. Specifically, S8797 with a 5-year PFS rate of 56%, S9313 with a 5-year DFS rate of 96%, S9415 with a median DFS of 8.5 years, and S9321 with a median PFS of 1.3 years, S9438 with a 2-year DFS rate of 30%, and S9008 with a median RFS of 20 months were identified as poorer candidates for a phase 2/3 design using OS as the primary endpoint and PFS as the phase 2 endpoint, and perhaps these populations are better addressed by separate phase 2 and phase 3 studies. It is possible that in these disease settings there are better endpoints that would alleviate this issue and allow for the use of a phase 2/3 design.

Table 3 summarizes the actual the number of events, sample size, percentage of actual accrual and the phase 2 decision for each power/type I error combination. In general the phase 2 decision was consistent across the range of error rates. Of the trials identified as poor candidates, two of the trials would have essentially gone to full accrual (S8797 and S9438) and the remaining 4 studies (S9313, S9415, S9321, and S9008) would have generally made it to at least 50% accrual before the phase 2 analysis, with S9313 accruing over around 2,000 patients and S9415 accruing around 1,000 patients by the analysis.

Table 3.

Results of phase 2 interim analysis in SWOG phase 3 trials

1 2 3 4 5 6 7 8 9

Study Final Power: 85% 85% 85% 90% 90% 90% 95% 95% 95%
Result Type I error: 10% 15% 20% 10% 15% 20% 10% 15% 20%
S8797 Positive (accrual goal : 264, actual size: 268)
Events 109 87 71 133 109 91 173 145 125
N 262 262 262 262 262 262 262 262 262
Accrual (%) 99% 99% 99% 99% 99% 99% 99% 99% 99%
Phase II stop No No No No No No No No No

S9008 Positive (accrual goal : 600, actual size: 606)
Events 97 78 64 119 97 82 155 130 112
N 263 221 202 316 263 227 366 324 302
Accrual (%) 44% 37% 34% 53% 44% 38% 61% 54% 50%
Phase II stop No No No No No No No No No

S9308 Positive (accrual goal : 440, actual size: 432)
Events 45 36 30 55 45 38 72 61 52
N 119 110 98 138 119 110 152 143 127
Accrual (%) 27% 25% 22% 31% 27% 25% 35% 33% 29%
Phase II stop Yes Yes Yes Yes Yes Yes Yes Yes Yes

S9313 Negative (accrual goal : 3240, actual size: 3176)
Events 50 40 33 61 50 42 79 67 57
N 1796 1598 1455 1957 1796 1618 2180 2050 1860
Accrual (%) 55% 49% 45% 60% 55% 50% 67% 63% 57%
Phase II stop No No No No No No No No No

S9321 Negative (accrual goal : 500, actual size: 575)
Events 70 56 46 86 70 59 112 94 81
N 205 174 166 234 205 184 298 253 227
Accrual (%) 41% 35% 33% 47% 41% 37% 60% 51% 45%
Phase II stop Yes No Yes Yes Yes Yes Yes No Yes

S9415 Negative (accrual goal : 1600, actual size: 1135)
Events 207 166 136 253 207 174 330 277 238
N 844 763 704 965 844 787 1004 1004 919
Accrual (%) 53% 48% 44% 60% 53% 49% 63% 63% 57%
Phase II stop Yes Yes Yes Yes Yes Yes Yes Yes Yes

S9420 Negative (accrual goal : 700, actual size: 730)
Events 59 47 39 72 59 49 94 79 68
N 135 114 105 148 135 122 186 161 141
Accrual (%) 19% 16% 15% 21% 19% 17% 27% 23% 20%
Phase II stop Yes Yes Yes Yes Yes Yes Yes Yes Yes

S9438 Negative (accrual goal : 206, actual size: 204)
Events 88 70 58 107 88 74 140 118 101
N 194 174 155 194 194 183 194 194 194
Accrual (%) 94% 84% 75% 94% 94% 89% 94% 94% 94%
Phase II stop Yes Yes Yes Yes Yes Yes Yes Yes Yes

S9509 Negative (accrual goal : 440, actual size: 432)
Events 45 36 29 55 45 38 71 60 51
N 129 118 110 147 129 119 164 152 140
Accrual (%) 29% 27% 25% 33% 29% 27% 37% 35% 32%
Phase II stop Yes No No Yes Yes No Yes Yes No

S9916 Positive (accrual goal : 744, actual size: 770)
Events 26 21 17 32 26 22 41 35 30
N 100 92 87 117 100 93 128 122 114
Accrual (%) 13% 12% 12% 16% 13% 13% 17% 16% 15%
Phase II stop No No No No No No No No No

S0001 Negative (accrual goal : 400, actual size: 183)
Events 47 38 31 58 47 40 76 64 55
N 65 63 54 79 65 63 108 94 73
Accrual (%) 16% 16% 14% 20% 16% 16% 27% 24% 18%
Phase II stop Yes No No Yes Yes No Yes Yes Yes

S0003 Negative (accrual goal : 550, actual size: 397)
Events 69 55 45 84 69 58 109 92 79
N 148 121 105 160 148 122 191 169 157
Accrual (%) 27% 22% 19% 29% 27% 22% 35% 31% 29%
Phase II stop Yes Yes Yes Yes Yes Yes Yes Yes Yes

S0023 Negative (accrual goal : 672, actual size: 284)
Events 45 36 30 55 45 38 72 61 52
N 109 92 86 136 109 93 160 140 130
Accrual (%) 16% 14% 13% 20% 16% 14% 24% 21% 19%
Phase II stop Yes Yes Yes Yes Yes Yes Yes Yes Yes

S0124 Negative (accrual goal : 680, actual size: 671)
Events 84 67 55 102 84 70 133 112 96
N 148 130 123 168 148 136 218 180 165
Accrual (%) 22% 19% 18% 25% 22% 20% 32% 26% 24%
Phase II stop No No No No No No No No No

S0205 Negative (accrual goal : 704, actual size: 766)
Events 84 67 55 102 84 70 133 112 96
N 173 157 146 200 173 161 239 211 191
Accrual (%) 25% 22% 21% 28% 25% 23% 34% 30% 27%
Phase II stop Yes Yes Yes Yes Yes Yes Yes Yes Yes

Of the positive trials 3 of 4 would have continued at the phase 2 analysis across all scenarios. S9308, the positive study that stopped at the phase 2 analysis was positive for both OS and PFS at the final analysis, with median PFS of 2 versus 4 months and median OS of 6 versus 8 months. The PFS HR used in the analysis was 2, which is likely too large. Evaluating this study with a HR of 1.6 (equivalent to a 1.2 month improvement), the study would have continued in all scenarios except in the phase 2 setting with 85% power and 20% type I error.

Of the negative trials, 10 of 11 would have stopped at the phase 2 analysis for the majority of scenarios. Interestingly, S0124 would have never stopped early, and at the final analysis while OS was not significantly different between the treatment arms, PFS was significantly different between the treatment arms. Results were mixed in S9509 and S0001 with the studies failing to stop for futility based on the evaluation with all scenarios with 20% type I error rates for S9509 and those with 85% and 90% power for S0001. In addition, both studies would have failed to stop with the 85% power, 20% type I error combination.

Because the phase 2 power has a large impact on the adjusted phase 2/3 power, our recommendation is to use at least 90% power for the phase 2 design. Then, the choice of the type I error will be based on both acceptability of the proportion of negative studies that will proceed to the phase 3 portion of the study and the sample size and timing of the phase 2 analysis. The evaluation of the SWOG studies indicates that a false positive rate of 20% may be too liberal.

Discussion

While this manuscript focuses upon the statistical properties of a new clinical trial design, it is also important to note that this design has already been used in clinical oncologic research. For example, SWOG S1203, an on-going study, employs early monitoring based on complete response while the primary outcome is event-free survival. In addition, coauthors on this paper, acting as consultants for Threshold  Pharmaceuticals, recommended this design for the conduct of a randomized clinical trial of doxorubicin versus the combination of doxorubicin and TH302, a prodrug of the DNA alkylator bromo-isophosphoramide mustard. The company reported that this design enabled them to raise sufficient capital to fund this registration trial.

The evaluation of SWOG studies presented in this paper was based on the proposed model for PFS and OS. However, we note that numerous authors have discussed modeling the relationship between PFS and OS. (8994) The proposed model used in this paper is just one such possible model. We find the model to be an intuitive one for the setting described. For the design of a phase 2/3 study, the study team should assess the disease setting and potential outcomes to be used and then determine which model is most approach for the design.

It is important to note that while a phase 2/3 design has a high likelihood of stopping at the phase 2 analysis under the null hypothesis, implementation of a phase 2/3 design does mean that the study sponsor and team are prepared for the study to continue to the phase 3 portion. In addition another possible downside to this approach is that a separate publication of the phase 2 study will not occur, as it is inappropriate to publish the results of interim analyses of an on-going study.

Conclusions

The phase 2/3 design has been suggested as an approach to speeding up drug development. While not applicable in all settings, use a of phase 2/3 design with PFS and OS, as described in this manuscript, could reduce the time to discovery not only in study time but also by removing the development time between a phase 2 and a phase 3 trial. Through careful consideration and thorough evaluation of design properties, substantial gains could occur using this approach.

Acknowledgments

We would like to thank Patricia Arlauskas, Harry Erba, Bruce Redman, and Manuel Valdevieso for their extensive work in performing the literature review and gathering the relevant data.

Financial Support: This investigation was supported in part by the following PHS Cooperative Agreement grant numbers awarded by the National Cancer Institute, DHHS: CA32102, CA38926, CA46441, CA105409, CA42777 and NIH grant CA090998.

Footnotes

Conflicts of Interest: None

References

  • 1.Zhang JJ, Zhang L, Chen H, Murgo A, Dodd LE, Pazdur R, et al. Assessment of audit methodologies for bias evaluation of tumor progression in oncology clinical trials. Clin Cancer Res. 2013;19 doi: 10.1158/1078-0432.CCR-12-3364. xx-xx. [DOI] [PubMed] [Google Scholar]
  • 2.Redman M, Crowley J. Small Randomized Trials. J Thorac Oncol. 2007;2:1–2. doi: 10.1097/JTO.0b013e31802c8d84. [DOI] [PubMed] [Google Scholar]
  • 3.Lara PN, Redman MW. The Hazards of Randomized Phase II Trials. Ann Oncol. 2012;23:7–9. doi: 10.1093/annonc/mdr567. [DOI] [PubMed] [Google Scholar]
  • 4.Storer BA. sequential phase II/III trial for binary outcomes. Stat Med. 1990;9:229–235. doi: 10.1002/sim.4780090305. [DOI] [PubMed] [Google Scholar]
  • 5.Inoue LYT, Thall PF, Berry DA. Seamlessly expanding a randomized phase II trial to phase III. Biometrics. 2002;58:823–831. doi: 10.1111/j.0006-341x.2002.00823.x. [DOI] [PubMed] [Google Scholar]
  • 6.Hunsberger S, Zhao Y, Simon R. A comparison of phase II study strategies. Clin Cancer Res. 2009;215:5950–5955. doi: 10.1158/1078-0432.CCR-08-3205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Parmar MKB, Barthel FMS, Sydes M. Speeding up the evaluation of new agents in cancer. JNCI. 2008;100:1204–1214. doi: 10.1093/jnci/djn267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Schaid DJ, Ingle JN, Wieand S, Ahmann DL. A design for phase II testing of anticancer agents within a phase III clinical trial. Control Clin Trials. 1988;9:107–118. doi: 10.1016/0197-2456(88)90032-3. [DOI] [PubMed] [Google Scholar]
  • 9.Stallard N. Group-sequential methods for adaptive seamless phase II/III clinical trials. J Biopharm Stat. 2011;21:787–801. doi: 10.1080/10543406.2011.551335. [DOI] [PubMed] [Google Scholar]
  • 10.Todd S, Stallard N. A new clinical trial design combining phases II and III: sequential designs with treatment selection and a change of endpoint. Drug Inf J. 2005;39:109–118. [Google Scholar]
  • 11.Korn EL, Freidlin B, Abrams J, Halabi S. Design issues in randomized phase II/III trials. J Clin Oncol. 2012;30:667–671. doi: 10.1200/JCO.2011.38.5732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Korn RL, Crowley JJ. Overview: progression-free survival as an endpoint in clinical trials with solid tumors. Clin Cancer Res. 2013;19 doi: 10.1158/1078-0432.CCR-12-2934. xx-xx. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Villaruz LC, Socinski MA. The clinical viewpoint: definitions, limitations of RECIST, practical considerations of measurement. Clin Cancer Res. 2013;19 doi: 10.1158/1078-0432.CCR-12-2935. xx-xx. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Rubinstein LV, Korn EL, Freidlin B, Hunsberger S, Ivy SP, Smith MA. Design issues of randomized phase II trials and a proposal for phase II screening trials. J Clin Oncol. 2005;23:199–206. doi: 10.1200/JCO.2005.01.149. [DOI] [PubMed] [Google Scholar]
  • 15.Ellenberg SS, Fleming TR, DeMets DL. Data Monitoring Committees in Clinical Trials: A Practical Perspective. New York: John Wiley and Sons; 2002. [Google Scholar]
  • 16.Fleming TR, Sharples K, McCall J, Moore A, Rodgers A, Stewart R. Maintaining Confidentiality of Interim Data to Enhance Trial Integrity and Credibility. Clinical Trials. 2008;5:157–167. doi: 10.1177/1740774508089459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Prentice RL. Surrogate endpoints in clinical trials: definition and operational criteria. Stat Med. 1989;8:431–440. doi: 10.1002/sim.4780080407. [DOI] [PubMed] [Google Scholar]
  • 18.Goldman B, LeBlanc M, Crowley J. Interim futility analysis with intermediate endpoints. Clin Trials. 2008;5:14–22. doi: 10.1177/1740774507086648. [DOI] [PubMed] [Google Scholar]
  • 19.Gandara DR, Lara PN, Jr., Natale R, Belani C. Progress in small-cell lung cancer: the lowest common denominator. J Clin Oncol. 2008;26:4236–4238. doi: 10.1200/JCO.2008.17.2692. [DOI] [PubMed] [Google Scholar]
  • 20.Paridaens R, Biganzoli L, Bruning P, Klijn JG, Gamucci T, Houston S, et al. Paclitaxel versus doxorubicin as first-line single-agent chemotherapy for metastatic breast cancer: a European Organization for Research and Treatment of Cancer randomized study with cross-over. J Clin Oncol. 2000;18:724–733. doi: 10.1200/JCO.2000.18.4.724. [DOI] [PubMed] [Google Scholar]
  • 21.Kaufman B, Mackey JR, Clemens MR, Bapsy PP, Vaid A, Wardley A, et al. Trastuzumab plus anastrozole versus anastrozole along for the treatment of postmenopausal women with human epidermal growth factor receptor 2-positive, hormone receptor-positive metastatic breast cancer: results from the randomized phase III TAnDEM study. J Clin Oncol. 2009;27:5529–5537. doi: 10.1200/JCO.2008.20.6847. [DOI] [PubMed] [Google Scholar]
  • 22.Johnston S, Pippen J, Jr., Pivot X, Lichinitser M, Sadeghi S, Dieras V, et al. Lapatinib combined with letrozole versus letrozole and placebo as first-line therapy for postmenopausal hormone receptor-positive metastatic breast cancer. J Clin Oncol. 2009;27:5538–5546. doi: 10.1200/JCO.2009.23.3734. [DOI] [PubMed] [Google Scholar]
  • 23.Robert N, Leyland-Jones B, Asmar L, Belt R, Ilegbodu D, Loesch D, et al. Randomized phase III study of trastuzumab, paclitaxel and carboplatin compared with trastuzumab and paclitaxel in women with HER-2-overexpressing metastatic breast cancer. J Clin Oncol. 2006;24:2786–2792. doi: 10.1200/JCO.2005.04.1764. [DOI] [PubMed] [Google Scholar]
  • 24.Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T, et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med. 2006;355:2733–2743. doi: 10.1056/NEJMoa064320. [DOI] [PubMed] [Google Scholar]
  • 25.Cameron D, Casey M, Oliva C, Newstat B, Imwalle B, Geyer CE. Lapatinib plus capecitabine in women with HER-2-positive advanced breast cancer: final survival analysis of a phase III randomized trial. Oncologist. 2010;15:924–934. doi: 10.1634/theoncologist.2009-0181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Blackwell KL, Burstein HJ, Storniolo AM, Rugo HS, Sledge G, Aktan G, et al. Randomized study of lapatinib alone or in combination with trastuzumab in women with ErbB2-positive, trastuzumab-refractory metastatic breast cancer. J Clin Oncol. 2010;28:1124–1130. doi: 10.1200/JCO.2008.21.4437. [DOI] [PubMed] [Google Scholar]
  • 27.Di Leo A, Gomez HL, Aziz Z, Zvirbule Z, Bines J, Arbushites MC, et al. Phase III, double-blind, randomized study comparing lapatinib plus paclitaxel with placebo plus paclitaxel as first-line treatment for metastatic breast cancer. J Clin Oncol. 2008;26:5544–5552. doi: 10.1200/JCO.2008.16.2578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Saltz LB, Cox JV, Blanke C, Rosen LS, Fehrenbacher L, Moore MJ, et al. Irinotecan plus fluorouracil and leucovorin for metastatic colorectal cancer. N Engl J Med. 2000;343:905–914. doi: 10.1056/NEJM200009283431302. [DOI] [PubMed] [Google Scholar]
  • 29.Douillard JY, Cunningham D, Roth AD, Navarro M, James RD, Karasek P, et al. Irinotecan combined with fluorouracil compared with fluorouracil alone as first-line treatment for metastatic colorectal cancer: a multicenter randomized trial. Lancet. 2000;355:1041–1047. doi: 10.1016/s0140-6736(00)02034-1. [DOI] [PubMed] [Google Scholar]
  • 30.de Gramont A, Figer A, Seymour M, Homerin M, Hmissi A, Cassidy J, et al. Leucovorin and fluorouracil with or without oxaliplatin as first-line treatment in advanced colorectal cancer. J Clin Oncol. 2000;18:2938–2947. doi: 10.1200/JCO.2000.18.16.2938. [DOI] [PubMed] [Google Scholar]
  • 31.Giacchetti S, Perpoint B, Zidani R, Le Bail N, Faggiuolo R, Focan C, et al. Phase III multicenter randomized trial of oxaliplatin added to chronomodulated fluorouracil-leucovorin as first-line treatment of metastatic colorectal cancer. J Clin Oncol. 18:136–147. doi: 10.1200/JCO.2000.18.1.136. [DOI] [PubMed] [Google Scholar]
  • 32.Goldberg RM, Sargent DJ, Morton RF, Fuchs CS, Ramanathan RK, Williamson SK, et al. A randomized controlled trial of fluorouracil plus leucovorin, irinotecan, andoxaliplatin combinations in patients with previously untreated metastatic colorectal cancer. J Clin Oncol. 22:23–30. doi: 10.1200/JCO.2004.09.046. [DOI] [PubMed] [Google Scholar]
  • 33.Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 350:2335–2342. doi: 10.1056/NEJMoa032691. [DOI] [PubMed] [Google Scholar]
  • 34.Amado RG, Wolf M, Peeters M, Van Cutsem E, Siena S, Freeman DJ, et al. Wild-type KRAS is required for panitumumab efficacy in patients with metatstatic colorectal cancer. J Clin Oncol. 26:1626–1634. doi: 10.1200/JCO.2007.14.7116. [DOI] [PubMed] [Google Scholar]
  • 35.Van Cutsem E, Peeters M, Siena S, Humblet Y, Hendlisz A, Neyns B, et al. Open-label phase III trial of panitumumab plus best supportive care compared with best supportive care alone in patients with chemotherapy-refractory metastatic colorectal cancer. J Clin Oncol. 2007;25:1658–1664. doi: 10.1200/JCO.2006.08.1620. [DOI] [PubMed] [Google Scholar]
  • 36.Peeters M, Price TJ, Cervantes A, Sobrero AF, Ducreux M, Hotko Y, et al. Randomized phase III study of panitumumab with fluorouracil, leucovorin, and irinotecan (FOLFIRI) compared with FOLFIRI alone as second-line treatment in patients with metastatic colorectal. J Clin Oncol. 2010;28:4706–4713. doi: 10.1200/JCO.2009.27.6055. [DOI] [PubMed] [Google Scholar]
  • 37.Douillard JY, Siena S, Cassidy J, Tabernero J, Burkes R, Barugel M, et al. Randomized, phase III trial of panitumumab with infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first-line treatment in patients with previously untreated metastatic colorectal cancer: the PRIME study. J Clin Oncol. 2010;28:4697–4705. doi: 10.1200/JCO.2009.27.4860. [DOI] [PubMed] [Google Scholar]
  • 38.Hecht JR, Mitchell E, Chidiac T, Scroggin C, Hagenstad C, Spigel D, et al. A randomized phase IIIB trial of chemotherapy, bevacizumab, and panitumumab compared with chemotherapy and bevacizumab alone for metastatic colorectal cancer. J Clin Oncol. 27:672–680. doi: 10.1200/JCO.2008.19.8135. [DOI] [PubMed] [Google Scholar]
  • 39.Van Cutsem E, Köhne CH, Hitre E, Zaluski J, Chang Chien CR, Makhson A, et al. Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med. 2009;360:1408–1417. doi: 10.1056/NEJMoa0805019. [DOI] [PubMed] [Google Scholar]
  • 40.Van Cutsem E, Lang I, Folprecht G, Nowacki M, Barone C, Shchepotin I, et al. Cetuximab plus FOLFIRI: final data from the CRYSTAL study on the association of KRAS and BRAF biomarker status with treatment outcome. J Clini Oncol. 2010;28:15s. (suppl; abstr 3570) [Google Scholar]
  • 41.Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol. 2010;28:1061–1068. doi: 10.1200/JCO.2009.23.9764. [DOI] [PubMed] [Google Scholar]
  • 42.Sternberg CN, Hawkins RE, Wagstaff J, Salman P, Mardiak J, Barrios CHC, et al. A randomized, double-blind phase III study (VEG105192) of pazopanib (paz) versus placebo (pbo) in patients with advanced/metastatic renal cell carcinoma (mRCC): updated safety results. J Clin Oncol. 2011;20(suppl 7) abstr 313. [Google Scholar]
  • 43.Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Oudard S, et al. Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma. J Clin Oncol. 2009;27:3584–3590. doi: 10.1200/JCO.2008.20.1293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Jonasch E, Corn P, Pagliaro LC, Warneke CL, Johnson MM, Tamboli P, et al. Upfront, randomized, phase 2 trial of sorafenib versus sorafenib and low-dose interferon alfa in patients with advanced renal cell carcinoma. Cancer. 2010;116:57–65. doi: 10.1002/cncr.24685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Escudier B, Szczylik C, Hutson TE, Demkow T, Staehler M, Rolland F, et al. Randomized phase II trial of first-line treatment with sorafenib versus interferon alfa-2a in patients with metastatic renal cell carcinoma. J Clin Oncol. 2009;27:1280–1289. doi: 10.1200/JCO.2008.19.3342. [DOI] [PubMed] [Google Scholar]
  • 46.Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med. 2007;356:125–134. doi: 10.1056/NEJMoa060655. [DOI] [PubMed] [Google Scholar]
  • 47.Escudier B, Bellmunt J, Négrier S, Bajetta E, Melichar B, Bracarda S, et al. Phase III trial of bevacizlumab plus interferon alfa-2a in patients with metastatic renal cell carcinoma (AVOREN): final analysis of overall survival. J Clin Oncol. 2010;28:2144–2150. doi: 10.1200/JCO.2009.26.7849. [DOI] [PubMed] [Google Scholar]
  • 48.Rini BI, Halabi S, Rosenberg JE, Stadler WM, Vaena DA, Archer L, et al. Phase III trial of bevacizumab plus interferon alfa versus interferon alfa monotherapy in patients with metastatic renal carcinoma: final results of CALBG 90206. J Clin Oncol. 2010;28:2137–2143. doi: 10.1200/JCO.2009.26.5561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Rini BI, Halabi S, Rosenberg JE, Stadler WM, Vaena DA, Ou SS, et al. Bevacizumab plus interferon alfa compared with interferon alfa monotherapy in patients with metastatic renal cell carcinoma: CALGB 90206. J Clin Oncol. 2008;26:5422–5428. doi: 10.1200/JCO.2008.16.9847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med. 2007;356:2271–2281. doi: 10.1056/NEJMoa066838. [DOI] [PubMed] [Google Scholar]
  • 51.Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, et al. Phase 3 trial of everolimus for metastatic renal cell carcinoma. Cancer. 2010;116:4256–4265. doi: 10.1002/cncr.25219. [DOI] [PubMed] [Google Scholar]
  • 52.Hallek M, Fischer K, Fingerle-Rowson G, Fink AM, Busch R, Mayer J, et al. Addition of rituximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukaemia: a randomized, open-label, phase 3 trial. Lancet. 2010;376:1164–1174. doi: 10.1016/S0140-6736(10)61381-5. [DOI] [PubMed] [Google Scholar]
  • 53.Hillmen P, Skotnicki AB, Robak T, Jaksic B, Dmoszynska A, Wu J, et al. Alemtuzumab compared with chlorambucil as first-line therapy for chronic lymphocytic leukemia. J Clin Oncol. 2007;25:5616–5623. doi: 10.1200/JCO.2007.12.9098. [DOI] [PubMed] [Google Scholar]
  • 54.Knauf WU, Lissitchkov T, Aldaoud A, Liberati AM, Loscertales J, Herbrecht R, et al. Bendamustine compared with chlorambucil in previously untreated patients with chronic lymphocytic leukaemia: updated results of a randomized phase III trial. Br J Haematol. 2012;159:67–77. doi: 10.1111/bjh.12000. [DOI] [PubMed] [Google Scholar]
  • 55.Rai KR, Peterson BL, Appelbaum FR, Kolitz J, Elias L, Shepherd L, et al. Fludarabine compared with chlorombucil as primary therapy for chronic lymphocytic leukemia. N Engl J Med. 2000;343:1750–1757. doi: 10.1056/NEJM200012143432402. [DOI] [PubMed] [Google Scholar]
  • 56.Wozniak AJ, Crowley JJ, Balcerzak SP, Weiss GR, Spiridonidis CH, Baker LH, et al. Randomized trial comparing cisplatin with cisplatin plus vinorelbine in the treatment of advanced non-small-cell lung cancer: a Southwest Oncology Group study. J Clin Oncol. 1998;16:2459–2465. doi: 10.1200/JCO.1998.16.7.2459. [DOI] [PubMed] [Google Scholar]
  • 57.Depierre A, Chastang C, Quoix E, Lebeau B, Blanchon F, Paillot N, et al. Vinorelbine versus vinorelbine plus cisplatin in advanced non-small cell lung cancer: a randomized trial. Ann Oncol. 1994;(1):37–42. doi: 10.1093/oxfordjournals.annonc.a058687. [DOI] [PubMed] [Google Scholar]
  • 58.Vogelzang NJ, Rusthoven JJ, Symanowski J, Denham C, Kaukel E, Ruffie P, et al. Phase III study of pemetrexed in combination with cisplatin versus cisplatin alone in patients with malignant pleural mesothelioma. J Clin Oncol. 2003;21:2636–2644. doi: 10.1200/JCO.2003.11.136. [DOI] [PubMed] [Google Scholar]
  • 59.Cohen MH, Johnson JR, Chen Y-F, Sridhara R, Pazdur R. FDA drug approval summary: erlotinib (tarceva) tablets. Oncologist. 2005;15:1344–1351. doi: 10.1634/theoncologist.10-7-461. [DOI] [PubMed] [Google Scholar]
  • 60.Inoue A, Kobayashi K, Maemondo M, Sugawara S, Oizumi S, Isobe H, et al. Updated overall survival results from a randomized phase III trial comparing gefitinib with carboplatin-paclitaxel for chemo-naïve non-small cell lung cancer with sensitive EGFR gene mutations (NEJ002) Ann Oncol. 2013;24:54–9. doi: 10.1093/annonc/mds214. [DOI] [PubMed] [Google Scholar]
  • 61.Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H, et al. Gefitinib or chemotherapy for non-small-cell lung cancer and mutated EGRF. N Engl J Med. 2010;362:2380–2388. doi: 10.1056/NEJMoa0909530. [DOI] [PubMed] [Google Scholar]
  • 62.Herbst RS, Giaccone G, Schiller JH, Natale RB, Miller V, Manegold C, et al. Gefitinib in combination with paclitaxel and carboplatin in advanced non-small cell lung cancer: a phase III trial – INTACT 2. J Clin Oncol. 22:785–794. doi: 10.1200/JCO.2004.07.215. [DOI] [PubMed] [Google Scholar]
  • 63.Giaccone G, Herbst RS, Manegold C, Scagliotti G, Rosell R, Miller V, et al. Gefitinib in combination with gemcitabine and cisplatin in advanced non-small-cell lung cancer: a phase III trial - INTACT 1. J Clin Oncol. 2004;22:777–784. doi: 10.1200/JCO.2004.08.001. [DOI] [PubMed] [Google Scholar]
  • 64.Kim ES, Hirsh V, Mok T, Socinski MA, Gervais R, Wu YL, et al. Gefitinib versus docetaxel in previously treated non-small-cell lung cancer (INTEREST): a randomized phase III trial. Lancet. 2008;372:1809–1818. doi: 10.1016/S0140-6736(08)61758-4. [DOI] [PubMed] [Google Scholar]
  • 65.Mitsudomi T, Morita S, Yatabe Y, Negoro S, Okamoto I, Tsurutani J, et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomized phase 3 trial. Lancet Oncol. 2010;11 doi: 10.1016/S1470-2045(09)70364-X. 121-8. [DOI] [PubMed] [Google Scholar]
  • 66.Fisher RI, LeBlanc M, Press OW, Maloney DG, Unger JM, Miller TP. New treatment options have changed the survival of patients with follicular lymphoma. J Clin Oncol. 2005;23:8447–8452. doi: 10.1200/JCO.2005.03.1674. [DOI] [PubMed] [Google Scholar]
  • 67.Richardson PG, Sonneveld P, Schuster M, Irwin D, Stadtmauer E, Facon T, et al. Extended follow-up of a phase 3 trial in relapsed multiple myeloma: final time-to-event results of the APEX trial. Blood. 2007;110(10):3557–3560. doi: 10.1182/blood-2006-08-036947. [DOI] [PubMed] [Google Scholar]
  • 68.Weber DM, Chen C, Niesvizky R, Wang M, Belch A, Stadtmauer EA, et al. Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America. N Engl J Med. 2007;357:2133–2142. doi: 10.1056/NEJMoa070596. [DOI] [PubMed] [Google Scholar]
  • 69.Orlowski RZ, Nagler A, Sonneveld P, Bladé J, Hajek R, Spencer A, et al. Randomized phase III study of pegylated liposomal doxorubicin plus bortezomib compared with bortezomib alone in relapsed or refractory multiple myeloma: combination therapy improves time to progression. J Clin Oncol. 2007;25:3892–3901. doi: 10.1200/JCO.2006.10.5460. [DOI] [PubMed] [Google Scholar]
  • 70.Gay F, Hayman SR, Lacy MQ, Buadi F, Gertz MA, Kumar S, et al. Lenalidomide plus dexamethasone versus thalidomide plus dexamethasone in newly diagnosed multiple myeloma: a comparative analysis of 411 patients. Blood. 2010;115:1343–1250. doi: 10.1182/blood-2009-08-239046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Gay F, Hayman S, Lacy MQ, Buadi F, Gertz MA, Kumar S, et al. Superiority of lenalidomide-dexamethasone versus thalidomide-dexamethasone as initial therapy for newly diagnosed multiple myeloma. Blood (Ash Annual Meeting Abstracts) 2009:114. Abstract 3884. [Google Scholar]
  • 72.Kapoor P, Rajkumar SV, Dispenzieri A, Gertz MA, Lacy MQ, Dingli D, et al. Melphalan and prednisone versus melphalan, prednisone and thalidomide for elderly and/or transplant ineligible patients with multiple myeloma: a meta-analysis. Leukemia. 2011;25:689–96. doi: 10.1038/leu.2010.313. [DOI] [PubMed] [Google Scholar]
  • 73.Petrylak DP, Tangen CM, Hussain MH, Lara PN, Jr., Jones JA, Taplin ME, et al. Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer. N Engl J Med. 2004;351:1513–1520. doi: 10.1056/NEJMoa041318. [DOI] [PubMed] [Google Scholar]
  • 74.de Bono JS, Oudard S, Ozguroglu M, Hansen S, Machiels JP, Kocak I, et al. Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomized open-label trial. Lancet. 2010;376:1147–54. doi: 10.1016/S0140-6736(10)61389-X. [DOI] [PubMed] [Google Scholar]
  • 75.Barlogie B, Kyle RA, Anderson KC, Greipp PR, Lazarus HM, Hurd DD, McCoy J, et al. Standard chemotherapy compared with high-dose chemoradiotherapy for multiple myeloma: final results of phase III US Intergroup Trial S9321. J Clin Oncol. 2006;24(6):929–936. doi: 10.1200/JCO.2005.04.5807. [DOI] [PubMed] [Google Scholar]
  • 76.Kelly K, Crowley J, Bunn PA, Jr., Presant CA, Grevstad PK, Moinpour CM, et al. Randomized phase III trial of paclitaxel plus carboplatin versus vinorelbine plus cisplatin in the treatment of patients with advanced non--small-cell lung cancer: a Southwest Oncology Group trial. J Clin Oncol. 2001;19:3210–3218. doi: 10.1200/JCO.2001.19.13.3210. [DOI] [PubMed] [Google Scholar]
  • 77.Kelly K, Chansky K, Gaspar LE, Albain KS, Jett J, Ung YC, et al. Phase III trial of maintenance gefitinib or placebo after concurrent chemoradiotherapy and docetaxel consolidation in inoperable stage III non-small-cell lung cancer: SWOG S0023. J Clin Oncol. 2008;26:2450–2456. doi: 10.1200/JCO.2007.14.4824. [DOI] [PubMed] [Google Scholar]
  • 78.Lara PN, Jr., Natale R, Crowley J, Lenz HJ, Redman MW, Carleton JE, et al. Phase III trial of irinotecan/cisplatin compared with etoposide/cisplatin in extensive-stage small-cell lung cancer: clinical and pharmacogenomic results from SWOG S0124. J Clin Oncol. 2009;27:2530–2535. doi: 10.1200/JCO.2008.20.1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Leichman CG, Benedetti JK, Zalupski MM, Hochster H, Shields AF, Lenz HJ, et al. Assessment of infusional 5-fluorouracil schedule and dose intensity: a Southwest Oncology Group and Eastern Cooperative Oncology Group study. Clin Colorectal Cancer. 2005;5:119–123. doi: 10.3816/ccc.2005.n.024. [DOI] [PubMed] [Google Scholar]
  • 80.Linden HM, Haskell CM, Green SJ, Osborne CK, Sledge GW, Jr., Shapiro CL, et al. Sequenced compared with simultaneous anthracycline and cyclophosphamide in high-risk stage I and II breast cancer: final analysis from INT-0137 (S9313) J Clin Oncol. 2007;25:656–661. doi: 10.1200/JCO.2006.07.0847. [DOI] [PubMed] [Google Scholar]
  • 81.Macdonald JS, Smalley SR, Benedetti J, Hundahl SA, Estes NC, Stemmermann GN, et al. Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Engl J Med. 2001;345:725–730. doi: 10.1056/NEJMoa010187. [DOI] [PubMed] [Google Scholar]
  • 82.Peters WA3rd, Liu PY, Barrett RJ2nd, Stock RJ, Monk BJ, Berek JS, et al. Concurrent chemotherapy and pelvic radiation therapy compared with pelvic radiation therapy alone as adjuvant therapy after radical surgery in high-risk early-stage cancer of the cervix. J Clin Oncol. 2000;18:1606–1613. doi: 10.1200/JCO.2000.18.8.1606. [DOI] [PubMed] [Google Scholar]
  • 83.Petrylak DP, Tangen CM, Hussain MH, Lara PN, Jr., Jones JA, Taplin ME, et al. Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer. N Engl J Med. 2004;351:1513–1520. doi: 10.1056/NEJMoa041318. [DOI] [PubMed] [Google Scholar]
  • 84.Philip PA, Benedetti J, Corless CL, Wong R, O’Reilly EM, Flynn PJ, et al. Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: Southwest Oncology Group-directed intergroup trial S0205. J Clin Oncol. 2010;28:3605–3610. doi: 10.1200/JCO.2009.25.7550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Poplin EA, Benedetti JK, Estes NC, Haller DG, Mayer RJ, Goldberg RM, et al. Phase III Southwest Oncology Group 9415/Intergroup 0153 randomized trial of fluorouracil, leucovorin, and levamisole versus fluorouracil continuous infusion and levamisole for adjuvant treatment of stage III and high-risk stage II colon cancer. J Clin Oncol. 2005;23:1819–1825. doi: 10.1200/JCO.2005.04.169. [DOI] [PubMed] [Google Scholar]
  • 86.Thompson JA, Fisher RI, Leblanc M, Forman SJ, Press OW, Unger JM, et al. Total body irradiation, etoposide, cyclophosphamide, and autologous peripheral blood stem-cell transplantation followed by randomization to therapy with interleukin-2 versus observation for patients with non-Hodgkin lymphoma: results of a phase 3 randomized trial by the Southwest Oncology Group (SWOG 9438) Blood. 2008;111:4048–4054. doi: 10.1182/blood-2007-09-111708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Wozniak AJ, Crowley JJ, Balcerzak SP, Weiss GR, Spiridonidis CH, Baker LH, et al. Randomized trial comparing cisplatin with cisplatin plus vinorelbine in the treatment of advanced non-small-cell lung cancer: a Southwest Oncology Group study. J Clin Oncol. 1998;16:2459–2465. doi: 10.1200/JCO.1998.16.7.2459. [DOI] [PubMed] [Google Scholar]
  • 88.Williamson SK, Crowley JJ, Lara PN, Jr., McCoy J, Lau DH, Tucker RW, et al. Phase III trial of paclitaxel plus carboplatin with or without tirapazamine in advanced non-small-cell lung cancer: Southwest Oncology Group Trial S0003. J Clin Oncol. 2005;23:9097–104. doi: 10.1200/JCO.2005.01.3771. [DOI] [PubMed] [Google Scholar]
  • 89.Buyse M, Burzykowski T, Carroll K, Michiels S, Sargent DJ, Miller LL, et al. Progression-free survival is a surrogate for survival in advanced colorectal cancer (2007) J Clin Oncol. 2007;25:5218–5224. doi: 10.1200/JCO.2007.11.8836. [DOI] [PubMed] [Google Scholar]
  • 90.Broglio KR, Berry DA. Detecting an overall survival benefit that is derived from progression-free survival. JNCI. 2009;101:1642–1649. doi: 10.1093/jnci/djp369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Fleming TR, Rothmann MD, Lu HL. Issues in using progression-free survival when evaluating oncology products. J Clin Oncol. 2009;27:2874–2880. doi: 10.1200/JCO.2008.20.4107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Saad ED, Katz A, Hoff PM, Buyse M. Progression-free survival as surrogate and as true end point: insights from the breast and colorectal cancer literature. Annals of oncology. 2010;21(1):7–12. doi: 10.1093/annonc/mdp523. [DOI] [PubMed] [Google Scholar]
  • 93.Oza AM, Castonguay V, Tsoref D, Diaz-Padilla I, Karakasis K, Mackay H, et al. Progression-free survival in advanced ovarian cancer: A Canadian review and expert panel perspective. Current Oncology. 2011;18(SUPPL. 2):S20–S27. [PMC free article] [PubMed] [Google Scholar]
  • 94.Hotta K, Suzuki E, Di Maio M, Chiodini P, Fujiwara Y, Takigawa N, et al. Progression-free survival and overall survival in phase III trials of molecular-targeted agents in advanced non-small-cell lung cancer. Lung Cancer. 2013;79:20–26. doi: 10.1016/j.lungcan.2012.10.007. [DOI] [PubMed] [Google Scholar]

RESOURCES