In the era of molecular oncology, patients still define a useful therapy as one that allows them to live longer and helps them to live better. Although patient outcomes have clearly improved as a result of randomized controlled trials (RCT), it is critical that contemporary trials retain the perspective of these fundamental patient-centered outcomes. Trends in study design, results, and interpretation of oncology RCTs from the past provide a useful framework in which to consider how the research community may approach trial design in the future. Although the RCT remains the standard for establishing efficacy, this article also considers how population-based outcome studies can provide insight into effectiveness of new therapies and explores how the results of RCTs translate into benefit in the general population. Clin Cancer Res; 16(24); 5963–71. ©2010 AACR.

Although patient outcomes have improved as a result of clinical trials conducted since the 1970s, in the contemporary era, patients still define a useful therapy as one that improves the quality, or increases the quantity, of survival. Despite considerable change in the treatment and outcome of patients with cancer, it is critical that investigators, clinicians, and policy makers not lose sight of these fundamental patient-centered outcomes. Furthermore, although the randomized controlled trial (RCT) remains the gold standard for establishing efficacy of new therapies, increased effort is required to understand how the results of RCTs influence clinical practice and to assess the benefit of new treatments in the general population. This article explores how the RCT in oncology has evolved over time and proposes issues that require consideration in the design and interpretation of future trials. The article also considers how population-based outcome studies can provide insight into the uptake of new medical therapies and explores how the results of RCTs translate into benefit in the “real world.”

A recent overview of 321 RCTs of systemic therapy in breast, colorectal, and non–small cell lung cancer (NSCLC), published between 1975 and 2004, provides insight into the methods, results, and outcomes of oncology trials over time (Table 1; ref. 1). RCTs in oncology have become larger and increasingly involve multiple international centers. Although the median sample size of RCTs has increased from 100 to 446 patients per study, the total duration of study accrual for these trials has remained relatively stable since the 1970s. During the study period, encouraging changes occurred in the statistical methodology and endpoints in RCTs. The primary endpoint of oncology RCTs has shifted away from response rate to survival and other time-to-event endpoints. The proportion of trials using intention-to-treat (ITT) analyses has also increased. Despite these improvements, in the most recent decade (1995 to 2004), one third of systemic therapy RCTs published in high-impact and widely read journals did not explicitly state the primary endpoint of the study and only half of trials report ITT analyses of all randomized patients.

Table 1.

Trends in study design and methodology of oncology randomized controlled trials 1975 to 2004

RCTs1975 to 19841985 to 19941995 to 2004
Number 47 107 167 
Disease site    
Breast 19 (40%) 53 (50%) 81 (49%) 
 NSCLC 23 (49%) 29 (27%) 39 (23%) 
 CRC 5 (11%) 25 (23%) 47 (28%) 
Setting    
 Palliative 34 (72%) 66 (62%) 101 (60%) 
 Adjuvant 13 (28%) 40 (37%) 61 (37%) 
 Neoadjuvant 0 (0%) 1 (1%) 5 (3%) 
Organization    
 Multicenter 26 (60%) 87 (85%) 158 (95%) 
 International 12 (26%) 30 (28%) 86 (52%) 
 Cooperative group 13 (28%) 59 (56%) 71 (43%) 
Source of RCT funding    
 Government 28 (60%) 66 (62%) 51 (31%) 
 Industry 2 (4%) 23 (23%) 95 (57%) 
 Foundation 7 (15%) 14 (13%) 24 (14%) 
 Cooperative group 12 (26%) 59 (55%) 75 (45%) 
 Other 1 (2%) 0 (0%) 2 (1%) 
Primary endpoint*    
 Time-to-event endpoints 11 (39%) 60 (72%) 125 (78%) 
 OS 6 (21%) 51 (61%) 82 (51%) 
 DFS 2 (7%) 5 (6%) 16 (10%) 
 RFS 3 (11%) 4 (5%) 8 (5%) 
 TTP 0 (0%) 0 (0%) 19 (12%) 
 RR 15 (54%) 21 (25%) 23 (14%) 
 Other 2 (7%) 2 (2%) 13 (8%) 
Study design    
 Median sample size 100 (47) 249 (107) 446 (167) 
 Median time for accrual 30 mo (22) 41 mo (83) 33 mo (153) 
 Median follow-up 30 mo (6) 55 mo (38) 47 mo (105) 
 Any ITT analysis 32 (70%) 93 (87%) 155 (93%) 
 ITT all randomized 15 (33%) 24 (22%) 91 (54%) 
 ITT eligible only 17 (37%) 69 (64%) 64 (38%) 
RCTs1975 to 19841985 to 19941995 to 2004
Number 47 107 167 
Disease site    
Breast 19 (40%) 53 (50%) 81 (49%) 
 NSCLC 23 (49%) 29 (27%) 39 (23%) 
 CRC 5 (11%) 25 (23%) 47 (28%) 
Setting    
 Palliative 34 (72%) 66 (62%) 101 (60%) 
 Adjuvant 13 (28%) 40 (37%) 61 (37%) 
 Neoadjuvant 0 (0%) 1 (1%) 5 (3%) 
Organization    
 Multicenter 26 (60%) 87 (85%) 158 (95%) 
 International 12 (26%) 30 (28%) 86 (52%) 
 Cooperative group 13 (28%) 59 (56%) 71 (43%) 
Source of RCT funding    
 Government 28 (60%) 66 (62%) 51 (31%) 
 Industry 2 (4%) 23 (23%) 95 (57%) 
 Foundation 7 (15%) 14 (13%) 24 (14%) 
 Cooperative group 12 (26%) 59 (55%) 75 (45%) 
 Other 1 (2%) 0 (0%) 2 (1%) 
Primary endpoint*    
 Time-to-event endpoints 11 (39%) 60 (72%) 125 (78%) 
 OS 6 (21%) 51 (61%) 82 (51%) 
 DFS 2 (7%) 5 (6%) 16 (10%) 
 RFS 3 (11%) 4 (5%) 8 (5%) 
 TTP 0 (0%) 0 (0%) 19 (12%) 
 RR 15 (54%) 21 (25%) 23 (14%) 
 Other 2 (7%) 2 (2%) 13 (8%) 
Study design    
 Median sample size 100 (47) 249 (107) 446 (167) 
 Median time for accrual 30 mo (22) 41 mo (83) 33 mo (153) 
 Median follow-up 30 mo (6) 55 mo (38) 47 mo (105) 
 Any ITT analysis 32 (70%) 93 (87%) 155 (93%) 
 ITT all randomized 15 (33%) 24 (22%) 91 (54%) 
 ITT eligible only 17 (37%) 69 (64%) 64 (38%) 

Abbreviations: CRC, colorectal cancer; RR, response rate.

*Data shown are for studies in which a primary endpoint was either explicitly identified or implied in the article. Time-to-events endpoints include overall survival (OS), disease-free survival (DFS), relapse-free survival (RFS), time to progression (TTP).

†The number of studies for which these data were available is shown in parentheses.

Data from Booth et al (1).

The funding of clinical trials has also changed remarkably over the past four decades. In the 1970s and early 1980s, government funding provided support for most RCTs in oncology, and industry was responsible for funding approximately 4%. In the most recent decade, more than half of RCTs (57%) were funded by industry, whereas the proportion of studies funded by government support has decreased substantially (1).

The concept of “benefit” and what constitutes a “positive” clinical trial is complex and involves variables from several interrelated domains. However, at least three important variables merit specific comment. The first is the magnitude of the benefit conferred by the new therapy: To what degree is the new drug better than the old drug, and what is that benefit (i.e., effect size)? The second concept is the level of statistical significance one can assign to the observed difference in outcome between treatment arms. The third variable that may influence whether clinicians consider a trial “positive” or “negative” is what the study authors say in their summary of the study results (i.e., author conclusions). Author conclusions themselves are highly complex but may take into account variables such as the effect size, P-value, quality of life data, treatment toxicity, and financial costs. Furthermore, other more subtle influences may lead to distorted presentation of study results or “spin.” In a recent overview of 72 RCTs with statistically nonsignificant results for the primary endpoint, Boutron and colleagues found evidence of “spin” in more than 40% of articles (2).

As shown in Table 2, several insightful findings have emerged from tracking the results and interpretation of oncology RCTs since the 1970s. First, the effect size (i.e., the relative benefit of new drug versus old drug) across individual trials has remained stable over time. Second, a considerably greater proportion of RCTs has results that are statistically significant, likely directly related to the large increase in sample size. Third, despite effect size remaining stable since the 1970s, authors of modern RCTs are considerably more likely to call their trial “positive” and strongly endorse the experimental arm. Although sponsorship status is not associated with increased effect size, industry funded RCTs are more likely to strongly endorse novel treatments. Multivariate analyses suggest that independent predictors of endorsing the experimental therapy are significant P-values for primary endpoint, time-to-event primary endpoint, industry sponsorship, and effect size (1).

Table 2.

What constitutes a “positive” randomized controlled trial? trends over time in results and interpretation of benefit in oncology randomized controlled trials 1975 to 2004

Results and Interpretation of RCTs1975 to 19841985 to 19941995 to 2004
1. Effect size: to what degree is the new therapy better than the standard of care? Median HR (95% CI) 1.4 (1.0–2.3) 1.2 (1.0–2.4) 1.2 (1.1–1.3) 
2. Statistical significance: is the observed magnitude of difference between arms statistically significant? P < 0.05 for primary endpoint 23% 30% 42% 
3. Author conclusions: what do the authors say in their summary of the trial results? Author strong endorsement of experimental arm* 31% 39% 49% 
Results and Interpretation of RCTs1975 to 19841985 to 19941995 to 2004
1. Effect size: to what degree is the new therapy better than the standard of care? Median HR (95% CI) 1.4 (1.0–2.3) 1.2 (1.0–2.4) 1.2 (1.1–1.3) 
2. Statistical significance: is the observed magnitude of difference between arms statistically significant? P < 0.05 for primary endpoint 23% 30% 42% 
3. Author conclusions: what do the authors say in their summary of the trial results? Author strong endorsement of experimental arm* 31% 39% 49% 

*Author conclusions were scored on the basis of a statement in the study abstract using a modified 7-point Likert Scale as described by Ridker and colleagues (63).

Data from Booth et al (1).

Trends in RCTs over the past four decades provide a framework for discussing aspects of trial design and interpretation that may be improved as we look to the next generation of clinical trials. Although an exhaustive overview of specific issues is beyond the scope of this article, this section explores common themes that emerge from related literature (Fig. 1).

Fig. 1.

Checklist for investigators, clinicians, and policy-makers who are evaluating patient-centered outcomes in randomized controlled trials.

Fig. 1.

Checklist for investigators, clinicians, and policy-makers who are evaluating patient-centered outcomes in randomized controlled trials.

Close modal

Appropriate design of clinical trials

As described elsewhere in this CCR FOCUS series by LoRusso and colleagues (3), one aspect that continues to plague oncology drug development is the inefficiency of the phase II to phase III transition. In an overview of 100 phase II trials with promising results presented at the American Society of Clinical Oncology (ASCO) 1995 to 1996, Berthold and colleagues found that one decade later, only 13 phase II studies had moved to the phase III setting (4). In a survey of promising contemporary phase II trials, the primary reasons for not moving forward related to limited financial and patient resources and inability to access the drug. In the design and implementation of oncology trials, careful consideration must be given to how valuable resources for clinical research (i.e., patients, drug, and financial) are used.

In the era of targeted cancer therapy, an important challenge to trial design is to identify which subset of patients is likely to benefit from a novel therapy on the basis of molecular characteristics at the tumor level. Given the vast number of molecular agents that have been studied in RCTs, the list of therapies that have conferred major benefit to patients is remarkably short (5, 6). It is likely that one of the reasons we have not observed more substantial improvements in outcome is that the treatment effect is being diluted by patients who do not express the molecular target of interest. RCTs in breast cancer have successfully included specific subsets of patients on the basis of tumor characteristics for many years, dating back to early trials of hormonal therapy (7–9), and more recently for trastuzumab (10). On the contrary, the development of bevacizumab for advanced colorectal and breast cancers provides a telling example of what may happen when an appropriate drug and tumor target relationship is not considered in determining the study population of interest. Although bevacizumab may offer meaningful benefit to a molecular subtype of patients with these diseases, to date the pivotal trials of this agent in breast and colorectal cancer have included unselected patients and have been associated with modest improvements in patient outcomes (11–13). The development of erlotinib in unselected populations of patients with advanced lung and pancreas cancer is another such example (14, 15). The benefit of these therapies in unselected populations is further reduced when one considers the substantial financial costs and toxicities associated with treatment. As discussed elsewhere in this CCR FOCUS series by Dalton and colleagues it is very likely that biomarkers will play an increasingly important role in the era of personalized medicine (16).

The increasing sample size of clinical trials (1) puts further strain on a system in which patient accrual is already suboptimal in many environments. As discussed in a recent report from the Institute of Medicine (17), no more than 5% of patients with cancer are entered onto a clinical trial and a considerable portion of RCTs do not meet accrual success (18). Although many of these trials may close for reasons other than accrual, it is possible that large sample sizes required by RCTs may impair the ability of investigators to answer clinically important questions.

Clinically relevant endpoints

The ultimate goal of all cancer research is to improve outcomes for our patients. Although outcomes have been traditionally evaluated using endpoints such as tumor size and length of overall survival, increasing emphasis is being placed on quality of life and other patient-reported outcomes. In recent years, important changes in the management of some diseases have been based primarily on improvements in these domains (19, 20). For this reason, it is critical that patient-centered endpoints be evaluated and interpreted with the same level of rigor as traditional endpoints.

In their overview of 112 RCTs in advanced cancer in which quality of life was evaluated (21), Joly and colleagues found that only 19% (21/112) established an a priori hypothesis with respect to quality of life and that only 21% (24/112) found a minimal difference in quality-of-life change score that showed any clinical benefit. Parallel to this is the increasing use of surrogates of overall survival such as progression-free or disease-free survival. Although these endpoints have been shown to correlate with improvement in quantity and/or quality of life for patients in select diseases (22–25), it is of concern that surrogate endpoints are increasingly used in diseases for which little validation exists (26–28).

One of the greatest challenges in interpreting the results of oncology RCTs is distinguishing results that are clinically important from those that are statistically significant (29, 30). With the remarkable increase in RCT sample size and the resulting change in statistical power, it has become possible to detect very small differences in outcomes between treatment arms, which are rendered appealing by virtue of a “significant” P-value. In fact, it is quite likely that therapies evaluated in older trials associated with effect sizes that we now would consider adequate [i.e., hazard ratio (HR) = 0.80] may have been abandoned because of greater expectations in the past. An illustrative example of this is shown in Fig. 2, which depicts survival curves from two trials of patients with advanced NSCLC. The older trial by Cellerino and colleagues (31) accrued patients in 1985 to 1988, whereas the more recent Eastern Cooperative Oncology Group (ECOG) 4599 trial by Sandler and colleagues (32) accrued from 2001 to 2004. Taking into account the different scales of the horizontal axis, on first review, the difference between treatment arms in each study appears comparable. Due to a substantially smaller sample size, the observed difference in the Cellerino trial was not significant (HR 0.78, P = 0.153), whereas the same effect size in ECOG 4599 was significant (HR 0.79, P = 0.003) and led to regulatory approval of bevacizumab for advanced NSCLC. However, if the Cellerino trial had been repeated with a sample size comparable to ECOG 4599, it is possible that the result would have reached statistical significance and may have changed practice instead of the experimental therapy being abandoned. In fact, later trials of cytotoxic chemotherapy among patients with NSCLC did, in fact, identify a statistically significant improvement in overall survival comparable with that reported by Cellerino and colleagues.

Fig. 2.

Survival curves from comparative RCTs in advanced NSCLC. Survival curves for RCTs led by Cellerino (A, accrual 1985 to 1988; ref. 25) and Sandler/ECOG 4599 (B, accrual 2001 to 2004; ref. 26). Although these two RCTs observed the same magnitude of difference between treatment arms, the difference in sample size and statistical power resulted in the older trial being considered “negative” and the contemporary trial being considered “positive.” If the trial by Cellerino had a sample size comparable to ECOG 4599, it is very likely that the observed difference would have been statistically significant. A, the Cellerino trial compared alternating chemotherapy versus best supportive care (BSC) and was powered to show an improvement in survival rate from 25 to 50% at 1 year (HR 0.5). To detect this improvement with 5% two-sided α and 80% power required 65 deaths. The trial enrolled 128 patients. Observed median survival was 8 months in the experimental group and 5 months in the control group (P = 0.153). In describing the survival curve, the authors state that “the graphic comparison of the two intervals clearly indicates no significant difference in survival between the treatments.” The observed HR for 1-year survival (estimated from the curve to be 33% and 25% for experimental and control groups, respectively) is 0.78. To have adequate power to detect this magnitude of difference would have required 492 deaths. Reprinted with permission from Cellerino et al. (31). Copyright © 2008 American Society of Clinical Oncology. All rights reserved. B, the Sandler trial (ECOG 4599) compared paclitaxel-carboplatin with bevacizumab (BPC) to paclitaxel-carboplatin (PC) alone and was powered to detect a HR for death of 0.80 with a one-sided α of 2.5% and 81% power. To detect this improvement required 650 deaths. The trial enrolled 878 patients. Observed median survival in the experimental group was 12 months and 10 months in the control group (HR 0.79, P = 0.003). Reproduced with permission from Sandler et al (32). Copyright © 2006 Massachusetts Medical Society. All rights reserved.

Fig. 2.

Survival curves from comparative RCTs in advanced NSCLC. Survival curves for RCTs led by Cellerino (A, accrual 1985 to 1988; ref. 25) and Sandler/ECOG 4599 (B, accrual 2001 to 2004; ref. 26). Although these two RCTs observed the same magnitude of difference between treatment arms, the difference in sample size and statistical power resulted in the older trial being considered “negative” and the contemporary trial being considered “positive.” If the trial by Cellerino had a sample size comparable to ECOG 4599, it is very likely that the observed difference would have been statistically significant. A, the Cellerino trial compared alternating chemotherapy versus best supportive care (BSC) and was powered to show an improvement in survival rate from 25 to 50% at 1 year (HR 0.5). To detect this improvement with 5% two-sided α and 80% power required 65 deaths. The trial enrolled 128 patients. Observed median survival was 8 months in the experimental group and 5 months in the control group (P = 0.153). In describing the survival curve, the authors state that “the graphic comparison of the two intervals clearly indicates no significant difference in survival between the treatments.” The observed HR for 1-year survival (estimated from the curve to be 33% and 25% for experimental and control groups, respectively) is 0.78. To have adequate power to detect this magnitude of difference would have required 492 deaths. Reprinted with permission from Cellerino et al. (31). Copyright © 2008 American Society of Clinical Oncology. All rights reserved. B, the Sandler trial (ECOG 4599) compared paclitaxel-carboplatin with bevacizumab (BPC) to paclitaxel-carboplatin (PC) alone and was powered to detect a HR for death of 0.80 with a one-sided α of 2.5% and 81% power. To detect this improvement required 650 deaths. The trial enrolled 878 patients. Observed median survival in the experimental group was 12 months and 10 months in the control group (HR 0.79, P = 0.003). Reproduced with permission from Sandler et al (32). Copyright © 2006 Massachusetts Medical Society. All rights reserved.

Close modal

Although small improvements should not by themselves change patient care, in some settings, these small steps may lead to substantial cumulative benefits over time. The sequential use of systemic therapy regimens in metastatic colon cancer is one such example in which results from cumulative RCTs have led to a major improvement in the outcomes of patients (33). In light of this paradox, Sobrero and Bruzzi have proposed a novel drug approval process in which drugs associated with modest benefit are granted provisional approval for further study and become eligible for full approval should a series of further studies show a cumulative and meaningful impact on patient outcomes (5). This new process may address some of the challenges of conducting definitive RCTs when the drug of interest has already received regulatory approval on “lesser” evidence.

Recognition is growing for the need to distinguish RCTs that are explanatory in nature from those that are pragmatic (34). Explanatory RCTs are conducted in tightly controlled settings with highly selected patients and are designed to explore the efficacy or biologic proof of concept of a novel therapy, often using short-term surrogate outcome measures. Results from these trials are generally used to inform further research rather than directly influence clinical practice. The initial randomized phase II trial of bevacizumab in advanced colorectal cancer (35) is one example of an explanatory trial. Despite the results showing improved survival in the experimental arm, the trial was designed and interpreted in the context of how to best inform subsequent studies of bevacizumab rather than directly influencing clinical practice. The pivotal RCT of erlotinib in advanced pancreatic cancer (15) is an example of an explanatory phase III trial; although the study met its primary endpoint, the results were presented in the context of informing further study rather than directly guiding patient care.

At the other end of the continuum, pragmatic trials test the effectiveness of an intervention in a setting that more closely resembles routine practice, with outcomes that have direct relevance to clinicians, patients, and policy makers. The results of pragmatic trials are to be used directly by those involved in providing patient care. MRC CR07/NCIC-CTG CO16 RCT (36) was a large pragmatic trial designed to evaluate the impact of preoperative radiotherapy on outcomes of patients with resectable rectal cancer. Patients were enrolled from 80 centers across five countries. Surgery was guided by local policy without central training. The study results showed a clear role for short-course radiotherapy among patients with rectal cancer in the general population. Identifying whether a RCT is pragmatic versus explanatory in nature may provide a framework for stakeholders in determining how the results of the study can and/or should be used.

Reporting of trials and avoidance of bias

As we strive to translate the results of oncology RCTs into clinical practice, it is essential that language be used that will allow clinicians and patients to clearly understand the potential benefit of a specific therapy. Elsewhere in this CCR FOCUS series, Smith and Hillner (37) provide a framework for having honest discussions with our patients. Saltz has recently proposed changes to common terminology used in the reporting of clinical trials (30). He emphasized the need to distinguish results that are only statistically significant from those that are clinically meaningful. He also suggested that commonly used phrases such as progression-free “survival,” reduction of the “risk of death,” and “tolerable” toxicity be reconsidered in an effort to improve clarity. Our own group recently described use of “clinical benefit” as an endpoint since it was originally described as a composite measure of pain, performance status, and weight among patients with pancreas cancer (38). Since the pivotal trial of gemcitabine in advanced pancreas cancer reported this outcome in 1997 (20), 71 trials reported in the Journal of Clinical Oncology have included a clinical benefit endpoint. Among these articles, 72% used “clinical benefit” to describe changes in objective tumor measurements (38). Whether such patients experience true clinical benefit depends on whether they also have improvement in the duration and/or quality of survival.

How and when the results of RCTs are disseminated is receiving increasing attention. It has become common for RCTs to terminate early on the basis of the results of interim analyses. Korn and colleagues recently reviewed 27 such trials conducted by National Cancer Institute (NCI) cooperative groups (39): Follow-up reports of these 27 trials suggest that the initial results and conclusions are preserved with longer follow-up. Termination based on a planned interim analysis is quite different to informal reporting of preliminary analyses at major cancer meetings. Preliminary data should not be made public for epidemiologic and statistical reasons (40, 41), which primarily relate to the statistical instability of early results and the potential for nonfinal analyses to misinform clinical practice. In a recent overview of 138 oncology RCTs published between 2000 and 2004, 44% of related conference abstracts used language to imply that the data were nonfinal and 63% of abstracts contained important data discordance when compared with the final publication (42). Furthermore, when compared with published articles, authors' conclusions were substantively discordant in 10% of abstracts. Accordingly, presentation of nonfinal results should be discouraged and clinicians should be cautious when interpreting immature data from clinical trials.

In the past decade, two sources of potential bias in oncology clinical trials have been described: publication bias (the preferential publication of RCTs with “positive results”; ref. 43) and sponsorship bias. It is hoped that publication bias will diminish with mandatory trial registration (44) and an increasing trend among major journals to publish negative trials. Although several groups have found that industry-sponsored studies are more likely to be positive than those not sponsored by industry, the reasons for this are likely to be complex and multifactorial (1, 45, 46). Possible explanations for this observation include: industry having a different threshold for determining whether to take a novel therapy to phase III study; differences in trial design and selection of the control arm; publication bias; and selective interpretation of trial results (2, 47). As consumers of medical literature, both clinicians and policy makers should bear publication and sponsorship biases in mind when appraising the evidence.

To better evaluate the impact of cancer therapies in the general population, it is necessary to look beyond the traditional clinical trial paradigm. Although the RCT offers excellent internal validity, because patients, physicians, and concurrent care in the general population may be very different from the controlled context of a clinical trial (48), it is essential that we not assume that the benefits shown in RCTs will be fully realized at the population level.

Recently, innovative study designs and access to large electronic databases have allowed investigators to evaluate the impact of new medical therapies in the “real world” (49, 50). These population-based outcome studies may provide insight into: uptake of new therapy, the impact of a change in practice and/or policy on outcomes, and the societal benefit of medical research. Furthermore, as discussed elsewhere in this CCR FOCUS series by Schnipper and colleagues, consideration of economic aspects of cancer care from a population perspective are urgently needed (51). Population-based studies also provide a mechanism to identify gaps in care following publication of a pivotal RCT and can facilitate targeted efforts in knowledge translation. Investigators can also learn about toxicities and outcomes associated with therapies when they are given to patient groups that are often underrepresented in RCTs (i.e., the elderly and those with other medical conditions). Important limitations to this study design include the possibility that changes in outcome at the population level may reflect confounding by other changes in treatment and/or case mix. However, given the unique insights they provide outside of the controlled environment of clinical trials, population-based outcome studies are becoming increasingly recognized as important in the generation and dissemination of medical evidence (50, 52–54). Two recent examples of such studies are described below.

In the past decade, pivotal RCTs have established clear roles for chemotherapy given concurrently with radiation in cervical cancer (55) and adjuvant chemotherapy in NSCLC (56, 57). Given the age and comorbidity of patients enrolled in clinical trials, concern was expressed about whether the RCT results were generalizable to the general population.

Following the NCI announcement recommending that “strong consideration should be given to adding chemotherapy to radiation therapy in the treatment of invasive cervical cancer” (55), Pearcey and colleagues undertook a population-based cohort study of the management and outcome of cervical cancer in Ontario (58). Using linked administrative and cancer registry databases, Pearcey and colleagues identified all cases of cervical cancer diagnosed in Ontario between 1992 and 2001 (N = 4,069). Prior to the NCI announcement, less than 10% of women treated with potentially curative radiotherapy received concurrent chemotherapy. Early in 1999, the use of concurrent chemotherapy increased rapidly, and from 1999 to 2001, more than 60% of such women received chemotherapy. The authors found that adoption of concurrent chemotherapy was associated with an 11% improvement in 3-year overall survival (P < 0.01). This survival benefit was virtually identical to that which would have been expected on the basis of the results of the clinical trials. Furthermore, the rate of hospitalizations during treatment did not increase, suggesting that the new treatment did not add substantial toxicity.

Using similar methodology, our group has recently explored uptake of adjuvant chemotherapy for NSCLC (59). Using the Ontario Cancer Registry linked to electronic records of treatment, we identified all 6,304 cases of NSCLC in Ontario that underwent surgical resection. During the study period, the proportion of surgical cases receiving adjuvant chemotherapy increased from 7 (2001 to 2003) to 31% (2004 to 2006). We also found a substantial improvement in survival among patients with resected NSCLC at the population level, consistent with the relevant clinical trials. Over the study period, there was no suggestion of major chemotherapy-related toxicity as measured by rates of hospitalization in the first 6 months after surgery.

These examples highlight how population-based outcome studies may complement knowledge gained from RCTs and serve to identify whether advances from clinical trials translate into meaningful benefit in the “real world”; this synergistic relationship is very much aligned with the principles of comparative effectiveness research (60). Furthermore, in an era when investment in health research is being increasingly scrutinized, these studies provide critical information related to the “research payback” of investments made in clinical trials (61).

Since the 1970s, large improvements have been made in the management and outcome of patients with cancer (62). The design and conduct of RCTs are responsible for many of these advances. As we look forward to the next generation of clinical trials, it will be critical to keep patient-centered outcomes at the forefront of trial design. Clear communication of study methodology and benefit of therapy is essential to convey to patients and other stakeholders the benefit of novel therapies. Finally, as we look to build on the advances made through RCTs, population-based outcome studies are needed to provide complementary data to understand how practice changes after publication of landmark RCTs and whether new medical therapies benefit patients in the “real world.”

No potential conflicts of interest were disclosed.

Drs. Ralph Meyer, Dongsheng Tu, Ian Tannock, Patricia LoRusso, and Steven Averbuch are gratefully acknowledged for thoughtful comments provided on an earlier version of this article.

C.M. Booth is supported as a Cancer Care Ontario Chair in Health Services Research.

1.
Booth
CM
,
Cescon
DW
,
Wang
L
,
Tannock
IF
,
Krzyzanowska
MK
. 
Evolution of the randomized controlled trial in oncology over three decades
.
J Clin Oncol
2008
;
26
:
5458
64
.
2.
Boutron
I
,
Dutton
S
,
Ravaud
P
,
Altman
DG
. 
Reporting and interpretation of randomized controlled trials with statistically nonsignificant results for primary outcomes
.
JAMA
2010
;
303
:
2058
64
.
3.
LoRusso
PM
,
Anderson
AB
,
Boerner
SA
,
Averbuch
SD
. 
Making the investigational oncology pipeline more efficient and effective – are we headed in the right direction
?
Clin Cancer Res
2010
;
16
:
5956
62
.
4.
Berthold
DR
,
Gulamhusein
A
,
Jackson
JI
,
Tannock
IF
. 
The transition from phase II to phase III studies
.
J Clin Oncol
2009
;
27
:
1150
1
.
5.
Sobrero
A
,
Bruzzi
P
. 
Incremental advance or seismic shift? The need to raise the bar of efficacy for drug approval
.
J Clin Oncol
2009
;
27
:
5868
73
.
6.
Fojo
T
,
Parkinson
DR
. 
Biologically targeted cancer therapy and marginal benefits: are we making too much of too little or are we achieving too little by giving too much
?
Clin Cancer Res
2010
;
16
:
5972
80
.
7.
Muss
HB
,
Case
LD
,
Atkins
JN
, et al
Tamoxifen versus high-dose oral medroxyprogesterone acetate as initial endocrine therapy for patients with metastatic breast cancer: a Piedmont Oncology Association study
.
J Clin Oncol
1994
;
12
:
1630
8
.
8.
Bonneterre
J
,
Thurlimann
B
,
Robertson
JF
, et al
Anastrozole versus tamoxifen as first-line therapy for advanced breast cancer in 668 postmenopausal women: results of the Tamoxifen or Arimidex Randomized Group Efficacy and Tolerability study
.
J Clin Oncol
2000
;
18
:
3748
57
.
9.
Nabholtz
JM
,
Buzdar
A
,
Pollak
M
, et al
Anastrozole is superior to tamoxifen as first-line therapy for advanced breast cancer in postmenopausal women: results of a North American multicenter randomized trial. Arimidex Study Group
.
J Clin Oncol
2000
;
18
:
3758
67
.
10.
Slamon
DJ
,
Leyland-Jones
B
,
Shak
S
, et al
Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2
.
N Engl J Med
2001
;
344
:
783
92
.
11.
Miles
DW
,
Chan
A
,
Dirix
LY
, et al
Phase III study of bevacizumab plus docetaxel compared with placebo plus docetaxel for the first-line treatment of human epidermal growth factor receptor 2-negative metastatic breast cancer
.
J Clin Oncol
2010
;
28
:
3239
47
.
12.
Robert
NJ
,
Dieras
V
,
Glaspy
J
. 
RIBBON 1: Randomized, double-blind, placebo-controlled phase 3 trial of chemotherapy with or without bevacizumab (B) for first-line treatment of HER-2 negative locally recurrent or metastatic breast cancer (MBC)
.
J Clin Oncol
2009
;
27
:
1005
.
13.
Cassidy
J
,
Clarke
S
,
Diaz-Rubio
E
, et al
Randomized phase III study of capecitabine plus oxaliplatin compared with fluorouracil/folinic acid plus oxaliplatin as first-line therapy for metastatic colorectal cancer
.
J Clin Oncol
2008
;
26
:
2006
12
.
14.
Shepherd
FA
,
Rodrigues
PJ
,
Ciuleanu
T
, et al
Erlotinib in previously treated non-small-cell lung cancer
.
N Engl J Med
2005
;
353
:
123
32
.
15.
Moore
MJ
,
Goldstein
D
,
Hamm
J
, et al
Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group
.
J Clin Oncol
2007
;
25
:
1960
6
.
16.
Dalton
WS
,
Sullivan
DM
,
Yeatman
TJ
,
Fenstermacher
DA
. 
The 2010 Health Care Reform Act: a potential opportunity to advance cancer research by taking cancer personally
.
Clin Cancer Res
2010
;
16
:
5987
96
.
17.
Institute of Medicine
. 
A national cancer clinical trials system for the 21st century: reinvigorating the NCI Cooperative Group Program
.
Washington
:
The National Academies Press
; 
2010
.
18.
Cheng
S
,
Dietrich
M
,
Finnigan
S
, et al
A sense of urgency: Evaluating the link between clinical trial development time and the accrual performance of CTEP-sponsored studies
.
J Clin Oncol
2009
;
27
:
CRA6509
.
19.
Tannock
IF
,
de Wit
R
,
Berry
WR
, et al
Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer
.
N Engl J Med
2004
;
351
:
1502
12
.
20.
Burris
HA
 III
,
Moore
MJ
,
Andersen
J
, et al
Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial
.
J Clin Oncol
1997
;
15
:
2403
13
.
21.
Joly
F
,
Vardy
J
,
Pintilie
M
,
Tannock
IF
. 
Quality of life and/or symptom control in randomized clinical trials for patients with advanced cancer
.
Ann Oncol
2007
;
18
:
1935
42
.
22.
Sargent
DJ
,
Wieand
HS
,
Haller
DG
, et al
Disease-free survival versus overall survival as a primary end point for adjuvant colon cancer studies: individual patient data from 20,898 patients on 18 randomized trials
.
J Clin Oncol
2005
;
23
:
8664
70
.
23.
Bast
RC
,
Thigpen
JT
,
Arbuck
SG
, et al
Clinical trial endpoints in ovarian cancer: report of an FDA/ASCO/AACR Public Workshop
.
Gynecol Oncol
2007
;
107
:
173
6
.
24.
Buyse
M
,
Thirion
P
,
Carlson
RW
,
Burzykowski
T
,
Molenberghs
G
,
Piedbois
P
. 
Relation between tumour response to first-line chemotherapy and survival in advanced colorectal cancer: a meta-analysis. Meta-Analysis Group in Cancer
.
Lancet
2000
;
356
:
373
8
.
25.
Paesmans
M
,
Sculier
JP
,
Libert
P
, et al
Response to chemotherapy has predictive value for further survival of patients with advanced non-small cell lung cancer: 10 years experience of the European Lung Cancer Working Party
.
Eur J Cancer
1997
;
33
:
2326
32
.
26.
Howell
A
,
Cuzick
J
,
Baum
M
, et al
Results of the ATAC (arimidex, tamoxifen, alone or in combination) trial after completion of 5 years' adjuvant treatment for breast cancer
.
Lancet
2005
;
365
:
60
2
.
27.
Ng
R
,
Pond
GR
,
Tang
PA
,
MacIntosh
PW
,
Siu
LL
,
Chen
EX
. 
Correlation of changes between 2-year disease-free survival and 5-year overall survival in adjuvant breast cancer trials from 1966 to 2006
.
Ann Oncol
2008
;
19
:
481
6
.
28.
Motzer
RJ
,
Hutson
TE
,
Tomczak
P
, et al
Sunitinib versus interferon alfa in metastatic renal-cell carcinoma
.
N Engl J Med
2007
;
356
:
115
24
.
29.
Fojo
T
,
Grady
C
. 
How much is life worth: cetuximab, non-small cell lung cancer, and the $440 billion question
.
J Natl Cancer Inst
2009
;
101
:
1044
8
.
30.
Saltz
LB
. 
Progress in cancer care: the hope, the hype, and the gap between reality and perception
.
J Clin Oncol
2008
;
26
:
5020
1
.
31.
Cellerino
R
,
Tummarello
D
,
Guidi
F
, et al
A randomized trial of alternating chemotherapy versus best supportive care in advanced non-small-cell lung cancer
.
J Clin Oncol
1991
;
9
:
1453
61
.
32.
Sandler
A
,
Gray
R
,
Perry
MC
, et al
Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer
.
N Engl J Med
2006
;
355
:
2542
50
.
33.
Meyerhardt
JA
,
Mayer
RJ
. 
Systemic therapy for colorectal cancer
.
N Engl J Med
2005
;
352
:
476
87
.
34.
Zwarenstein
M
,
Treweek
S
. 
What kind of randomized trials do we need
?
CMAJ
2009
;
180
:
998
1000
.
35.
Kabbinavar
FF
,
Schulz
J
,
McCleod
M
, et al
Addition of bevacizumab to bolus fluorouracil and leucovorin in first-line metastatic colorectal cancer: results of a randomized phase II trial
.
J Clin Oncol
2005
;
23
:
3697
705
.
36.
Sebag-Montefiore
D
,
Stephens
RJ
,
Steele
R
, et al
Preoperative radiotherapy versus selective postoperative chemoradiotherapy in patients with rectal cancer (MRC CR07 and NCIC-CTG C016): a multicentre, randomised trial
.
Lancet
2009
;
373
:
811
20
.
37.
Smith
TJ
,
Hillner
BE
. 
Explaining marginal benefits to patients, when “marginal” means additional but not necessarily small
.
Clin Cancer Res
2010
;
16
:
5981
6
.
38.
Ohorodnyk
P
,
Eisenhauer
EA
,
Booth
CM
. 
Clinical benefit in oncology trials: is this a patient-centred or tumour-centred end-point
?
Eur J Cancer
2009
;
45
:
2249
52
.
39.
Korn
EL
,
Freidlin
B
,
Mooney
M
. 
Stopping or reporting early for positive results in randomized clinical trials: the National Cancer Institute Cooperative Group experience from 1990 to 2005
.
J Clin Oncol
2009
;
27
:
1712
21
.
40.
Fleming
TR
,
Ellenberg
S
,
DeMets
DL
. 
Monitoring clinical trials: issues and controversies regarding confidentiality
.
Stat Med
2002
;
21
:
2843
51
.
41.
Piantadosi
S
. 
Rigor in monitoring clinical trials is ethical
.
J Clin Oncol
2008
;
26
:
683
5
.
42.
Booth
CM
,
Le
MA
,
Ding
K
, et al
Presentation of nonfinal results of randomized controlled trials at major oncology meetings
.
J Clin Oncol
2009
;
27
:
3938
44
.
43.
Krzyzanowska
MK
,
Pintilie
M
,
Tannock
IF
. 
Factors associated with failure to publish large randomized trials presented at an oncology meeting
.
JAMA
2003
;
290
:
495
501
.
44.
De Angelis
C
,
Drazen
JM
,
Frizelle
FA
, et al
Clinical trial registration: a statement from the International Committee of Medical Journal Editors
.
N Engl J Med
2004
;
351
:
1250
1
.
45.
Djulbegovic
B
,
Lacevic
M
,
Cantor
A
, et al
The uncertainty principle and industry-sponsored research
.
Lancet
2000
;
356
:
635
8
.
46.
Peppercorn
J
,
Blood
E
,
Winer
E
,
Partridge
A
. 
Association between pharmaceutical involvement and outcomes in breast cancer clinical trials
.
Cancer
2007
;
109
:
1239
46
.
47.
Lexchin
J
,
Bero
LA
,
Djulbegovic
B
,
Clark
O
. 
Pharmaceutical industry sponsorship and research outcome and quality: systematic review
.
BMJ
2003
;
326
:
1167
70
.
48.
Meyer
RM
. 
Generalizing the results of cancer clinical trials
.
J Clin Oncol
2010
;
28
:
187
9
.
49.
Wen
SW
,
Kramer
MS
. 
Uses of ecologic studies in the assessment of intended treatment effects
.
J Clin Epidemiol
1999
;
52
:
7
12
.
50.
Booth
CM
,
Mackillop
WJ
. 
Translating new medical therapies into societal benefit: the role of population-based outcome studies
.
JAMA
2008
;
300
:
2177
9
.
51.
Schnipper
LE
,
Meropol
NJ
,
Brock
DW
. 
Value and cancer care: towards an equitable future
.
Clin Cancer Res
2010
;
16
.:
6004
8
52.
Vlahakes
GJ
. 
The value of phase 4 clinical testing
.
N Engl J Med
2006
;
354
:
413
5
.
53.
Avorn
J
. 
In defense of pharmacoepidemiology–embracing the yin and yang of drug research
.
N Engl J Med
2007
;
357
:
2219
21
.
54.
Juurlink
DN
. 
Rosiglitazone and the case for safety over certainty
.
JAMA
2010
;
304
:
469
71
.
55.
National Cancer Institute
.
Bethesda (MD)
:
National Cancer Institute
NCI issues clinical announcement on cervical cancer: chemotherapy plus radiation improves survival
; 
1999
.
Available from
: www cancer gov/newscenter/cervicalcancer.
56.
Winton
T
,
Livingston
R
,
Johnson
D
, et al
Vinorelbine plus cisplatin vs. observation in resected non-small-cell lung cancer
.
N Engl J Med
2005
;
352
:
2589
97
.
57.
Douillard
JY
,
Rosell
R
,
De Lena
M
, et al
Adjuvant vinorelbine plus cisplatin versus observation in patients with completely resected stage IB-IIIA non-small-cell lung cancer (Adjuvant Navelbine International Trialist Association [ANITA]): a randomised controlled trial
.
Lancet Oncol
2006
;
7
:
719
27
.
58.
Pearcey
R
,
Miao
Q
,
Kong
W
,
Zhang-Salomons
J
,
Mackillop
WJ
. 
Impact of adoption of chemoradiotherapy on the outcome of cervical cancer in Ontario: results of a population-based cohort study
.
J Clin Oncol
2007
;
25
:
2383
8
.
59.
Booth
CM
,
Shepherd
FA
,
Peng
Y
, et al
Adoption of adjuvant chemotherapy for non-small-cell lung cancer: a population-based outcomes study
.
J Clin Oncol
2010
;
28
:
3472
8
.
60.
Conway
PH
,
Clancy
C
. 
Comparative-effectiveness research–implications of the Federal Coordinating Council's report
.
N Engl J Med
2009
;
361
:
328
30
.
61.
Meyer
RM
. 
How to optimally measure the impact of health research funding in clinical research.In: Making an impact: a preferred framework and indicators to measure returns on investment in health research
.
Ottawa: Canadian Academy of Health Sciences
; 
2007
;
A2
A20
.
62.
American Cancer Society
. 
Cancer facts & figures 2008
.
Atlanta
:
American Cancer Society
; 
2008
.
63.
Ridker
PM
,
Torres
J
. 
Reported outcomes in major cardiovascular clinical trials funded by for-profit and not-for-profit organizations: 2000–2005
.
JAMA
2006
;
295
:
2270
4
.