Download Phase III Trials of Targeted Anti-cancer Therapies: redesigning the

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
no text concepts found
Transcript
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Phase III Trials of Targeted Anti-cancer Therapies: redesigning the
concept
Alberto Ocana, Eitan Amir, Francisco Vera-Badillo, Bostjan Seruga and Ian F. Tannock
From the Division of Medical Oncology and Hematology, Princess Margaret Hospital and
the University of Toronto, Toronto, Canada
Word count: Abstract 123, Text 3114
Keywords: phase III trials, adaptive design, anticancer-therapies, targeted agents, biomarkers
Running title: Phase III Trials of Targeted Anti-Cancer Therapies
Correspondence:
Ian F Tannock MD, PhD, DSc
Division of Medical Oncology and Hematology,
Princess Margaret Hospital,
610 University Avenue,
Toronto, ON M5G 2 M9,
Canada
T:
416-946-2245
F:
416-946-4563
E:
[email protected]
1
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Abstract
Randomized phase III trials provide the gold-standard evidence for the approval of new
drugs: an experimental treatment is compared with the current standard of care to identify
clinically relevant differences in a predefined endpoint. However, there are several problems
relating to the current role of phase III trials in drug development including the limited
clinical benefit observed for some approved agents, the necessity for large trials to detect
these differences, the inability of such trials to identify rare but important toxicities, and high
cost. The design of phase III trials evaluating drug combinations, and of those including
biomarkers, presents additional challenges. Here, we review these problems and suggest that
phase III trials with adaptive designs in selected prescreened populations could reduce these
limitations.
2
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Introduction
Phase III randomized clinical trials (RCTs) have been used to generate evidence in support of
the approval of most new agents in the treatment of cancer (1, 2). In phase III studies an
experimental treatment is compared with the standard of care to identify relevant differences
in predefined endpoints. In cancer drug development, these are usually time to event
endpoints such as overall (OS), progression-free (PFS) or disease-free survival (DFS) (1, 2).
If the magnitude of difference is statistically significant, the experimental treatment is usually
approved by regulatory authorities, and can then be incorporated into the therapeutic
armamentarium (1).
In the last decade, numerous targeted therapies have been evaluated in clinical trials and
many have been approved for the treatment of cancer. For most of these agents, phase III
trials were undertaken to detect significant differences in a predefined endpoint. However, for
a small selected group, impressive results observed in early studies led to approval without
the need to perform a randomized study (reviewed by Tsimberidou et al (3)).
The limited activity of some targeted agents, the necessity of large phase III trials to
demonstrate significant differences in endpoint, and the high cost at which they are marketed,
have raised questions about the way these agents are developed, including the necessity of
conducting phase III trials to demonstrate improved outcomes compared to standard
treatment (4-6). In the present article we review the role of phase III trials in the
development of targeted therapies, identify problems associated with them, and propose ideas
to improve the process of drug development. We suggest that modifications to the design of
phase II-III trials including adaptive models in a preselected screened population could help
to circumvent some of these problems (4, 5).
3
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Problems associated with drug development: role of randomized trials
Limited clinical benefit in large RCTs
The magnitude of difference in clinical benefit between experimental and standard treatment
is the key factor in convincing regulatory agencies, and some clinicians, of the utility of a
given drug. Although there is no consensus as to what should be considered a sufficient
magnitude of benefit from use of a new agent to recommend its adoption, for most
oncologists a gain in survival of at least 3-4 months for patients with incurable metastatic
disease is clinically important, especially if toxicity is acceptable and no deterioration in
quality of life is observed (6). However, there are examples of targeted agents that have been
approved by regulatory agencies with very limited improvement in absolute benefit: an
example was the approval of erlotinib in pancreatic cancer with an improvement in median
survival of around 10 days (see table 1) (7), a difference that was statistically significant but
less than pre-specified in the protocol as clinically important. A positive study should not be
based only on a statistical test alone but also on an improvement in a clinically relevant
endpoint that will translate into substantial patient benefit (6). This benefit should be
described in absolute terms since relatively impressive reductions in a hazard ratio (i.e. in
relative benefit) may translate into very small differences in an absolute measure such as
median survival.
Clinical trials have become larger with time (8), and large sample sizes are often used in
clinical trials to detect small benefits in predefined endpoints which meet conventional levels
of statistical significance and allow drug registration. For example, studies evaluating
erlotinib and bevacizumab for non-small cell lung cancer used large sample sizes to detect
small but statistically significant differences in outcome (9, 10). Similarly the recent approval
4
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
of Ziv-aflibercept in metastatic colon cancer was based on a clinical trial involving 1226
patients where the difference in median OS was only 6 weeks (11).
Predicting a successful phase III trial from phase II data
The completion of a randomized phase II trial with positive results does not guarantee a
subsequent positive phase III study. This may be due to more stringent selection of patients in
phase II studies, the use of different endpoints in phase II and III studies (e.g. PFS in the
phase II and OS in the phase III),lack of understanding how they relate to each other, low
statistical power in smaller phase II studies, or simply regression to the mean. Estimation of
the sample size for a phase III trial will depend on the magnitude of benefit expected, and
will be influenced by that observed in phase II trials. Ideally however the sample size should
not be set by the likelihood of obtaining an effect that is statistically significant, but by
detecting or ruling out one that is clinically important.
Are RCTs sufficient to determine toxicity?
Phase III trials should be designed to obtain information about toxicities related to the
investigational treatment as well as benefit. Phase III trials tend to capture frequent but mild
toxicity and sometimes less frequent but more severe toxicities not identified in previous
phase I and II studies (12). However, many phase III trials fail to capture this information
adequately. Around 40% of fatal adverse drug reactions (ADR) of approved targeted agents
reported in updated drug labels were not described in any published report of the phase III
trial(s) used for the approval of that drug (13): thus phase III trials are not sufficiently
powered to detect infrequent but serious toxicities of targeted agents, or those that may occur
after completion of the trial (14). In addition, phase III trials have limited ability to detect
toxicity in vulnerable subgroups of patients, who are often excluded from phase III studies
but may receive the drug once it is approved. Furthermore, our group has reviewed data using
5
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
meta-analyses, which show that newly approved anticancer drugs are associated with
increased drug-induced morbidity and mortality when compared with the standard treatment
received by control groups. These toxicities became evident only when data were pooled
from multiple RCTs (15). We do not suggest enlarging the sample size of RCTs to increase
their probability of demonstrating less common types of toxicity, but we do recommend strict
requirements for reporting of toxicity as a condition of marketing approval.
Pitfalls in the design of studies with drug combinations
Although the design of studies with drug combinations should be based on preclinical data to
support an interaction leading to improved therapeutic index, a recent analysis from our
group shows that to be uncommon: there is rarely preclinical information to suggest a
synergistic interaction between the agents evaluated in phase II trials (16). Since the activity
of targeted drugs as single agents has been modest, most of them have been developed in
association with standard chemotherapy, and they have usually been given concomitantly.
Scheduling of a primarily cytostatic targeted agent to be given concurrently with cycledependent cytotoxic chemotherapy makes little biological sense, since chemotherapy will be
less active if cells are put out of cycle by the targeted agent, although combined activity
against multiple pathways might overcome this antagonistic effect. Scheduling the targeted
agent between cycles of chemotherapy to inhibit tumor cell repopulation could be an option
to improve therapeutic outcome (17, 18). This approach has not been explored adequately in
clinical trials.
Phase I and early phase II trials provide opportunities to investigate dose and schedule for a
drug combination before launching a large randomized phase II or a phase III trial, but this is
often not undertaken. The FDA's Critical Path Initiative addresses the limited foundation of
early drug development studies (19). For example a recent phase IIB trial involving 229
6
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
patients with HER2 negative breast cancer evaluated the combination of sorafenib and
capecitabine compared with capecitabine alone; the combination showed unacceptable
toxicity that could have been detected in a smaller study (20) . Similarly the phase III trial
that compared standard chemotherapy (AC or paclitaxel) versus standard chemotherapy with
trastuzumab in metastatic breast cancer showed unacceptable toxicity in the AC-containing
arm, a combination that was never evaluated in a phase I or phase II trial (21). These
examples highlight the importance for the re-design of phase II-III trials with drug
combinations to facilitate the early identification of toxic combinations and to select the most
appropriate dose and schedule to obtain the best balance between clinical benefit and toxicity.
Schedule is as important as dose, as has been demonstrated recently when evaluating
different schedules of sunitinib in renal cell carcinoma (22). The design of phase II trials to
find the best schedule is challenging as resources are not usually available to evaluate various
schedules, so that one schedule is chosen with limited prior information. However, there
could be greater emphasis on dose scheduling in preclinical studies that could be refined in
the phase I trials.
Cost associated with development of targeted drugs
Recently approved drugs are marketed at a high price despite many of them having only
modest activity; thus many new drugs do not fall within a range of cost-effectiveness used for
evaluating other health interventions. In the approval process of a drug by some regulatory
authorities, market price and cost-effectiveness are not taken into consideration. There are
concerns about long-term affordability of new anticancer drugs for both patients and
healthcare providers (including public health systems and private insurers), and health
providers may not be willing to fund the costs of drugs if they do not meet standards of costeffectiveness (23). What is considered cost-effective differs between countries (24). For
7
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
developed countries up to $100,000 is often considered a reasonable maximum cost per
quality adjusted life year gained (24-27), but it will be far less in developing countries.
Renal cell carcinoma is one of the solid tumors where several new targeted agents have been
approved. Table 2 shows prices associated with each new treatment, including those for the
control and experimental arms of the pivotal trials, in addition to the clinical benefit observed
in the predefined endpoints and estimates of the incremental cost per life-year (or
progression-free if OS is not available) gained. Some of the new drugs do not fall within the
above standard of cost-benefit. One of the reasons proposed for the high market price is the
high cost of the drug development process and particularly that associated with the design of
large phase III trials.
Can adaptive phase II-III trial designs in a molecularly prescreened population help to
resolve these problems?
An adaptive trial uses data obtained while the trial is ongoing to modify the course of the trial
(28, 29). As suggested by the FDA and EMA (30), all potential adaptations should be
preplanned and registered before the trial is initiated. Examples of adaptive measures include
early stopping rules in case of lack of efficacy or unacceptable toxicity, adapting doses or
schedules that will lead to a more efficient benefit-toxicity relationship, stopping arms in a
multi-arm trial, changing accrual, selection and/or order of primary and secondary end-points,
or modification of concomitant treatments (28, 29, 30). In addition, adaptive trials can use
outcome-adaptive randomization in which the ratio of patients randomly assigned to the
experimental arm versus the control arm changes over time from the standard 1:1 to increase
the proportion of patients randomized to the arm that is doing better (28).
There are advantages to designing a study with various arms, including those with different
dose and schedule of a combination. With emerging (albeit imperfect) evidence that one arm
8
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
of the phase II-III trial is superior (i.e. has less toxicity and more activity in a predefined
interim analysis) - increased accrual to that arm can augment the statistical power to detect a
relevant magnitude of clinical benefit. An example of this multi-arm approach is the
STAMPEDE trial in prostate cancer (31): an increased benefit in biochemical (i.e. PSA)
response or PFS as an interim endpoint is used to support increased accrual to the selected
arms while maintaining OS as the final endpoint. Accrual to a phase III trial can be designed
to identify uncommon but serious toxicities or even to increase the accrual for a specific
vulnerable population.
The incorporation of several arms in a randomized trial with different doses and/or schedules
can facilitate identification of the best ratio of benefit to toxicity. This approach could have
been used to identify the best dose and schedule for the combination of sorafenib and
capecitabine in HER2 negative metastatic breast cancer patients (20). In contrast, the large
phase IIB study concluded that the combination was clinically active but toxic and another
trial was necessary to evaluate the combination using lower doses (20). An adaptive design
would have saved time and resources as early stopping rules would have led to dropping of
toxic or ineffective arms.
Although adaptive clinical trials can be more expensive initially as they may include more
scenarios and treatment arms, their ability to prevent the undertaking of further clinical
studies can reduce the total cost of drug development.
Incorporation of biomarkers in adaptive designs
The incorporation of biomarkers in phase III studies should be exploratory unless a validated
marker is identified and it should then be evaluated (quantified) for all included patients. The
incorporation of biomarkers in studies of approved drugs in the last decade is summarized in
table 3. We reviewed previously the magnitude of relative benefit in phase III trials used for
9
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
the approval of new drugs: although all studies were powered to detect about a 20-30%
reduction in relative risk, drugs designed to interact with a specific target, and especially
those that used predictive biomarkers, were able to produce the highest relative improvement
in OS and PFS (32) (figure 1). Thus the sample size needed to detect this difference was
smaller for these studies (32). Adaptive phase III trials using biomarkers and a preselected
population may not need a large sample to detect a relevant clinical benefit, and they can
have early stopping rules in case the study arm meets the predefined magnitude of benefit.
The above features can be included in a continuous phase II-III study design (figure 2). This
approach would exclude the majority of single arm trials that have no intention to proceed to
a phase III trial; this is a positive feature as most such trials represent a waste of resources
(33). In the phase II part, different arms can be evaluated but only the arm with signs of
major activity and acceptable toxicity will be included in the final phase III part that will be
compared with standard treatment. An example of this approach is the I-SPY2 trial in breast
cancer (34). The phase II part is exploratory and gives information about how to design the
phase III and what might be the expected magnitude of benefit and its uncertainty. In
addition, the endpoint for the phase II part could be a surrogate for the endpoint used in the
phase III trial. However, this assumption is often not justified: for example, there is rather
poor correlation between improvements in PFS and in OS. The total number of patients to be
included can also be higher as they will be combined with those included in the phase II part,
so the identification of a meaningful ratio of clinical benefit to toxicity will be easier (figure
2). This approach can reduce time and cost. An example will be a multi-arm trial in
metastatic breast cancer in which a specific experimental treatment arm shows an increase in
PFS of more than 6-8 months, like the results recently reported with pertuzumab in HER2positive breast cancer that has lead to FDA approval of this drug (35). The preliminary result
10
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
could give confidence to continue or increase accrual in that arm and stop the other
experimental arms, and aim for an improvement in OS of at least 3-4 months.
The design of phase II-III trials in a molecularly prescreened population can also help to
overcome some of the problems described earlier. Impressive clinical activity observed in a
phase I-II trial in a subgroup of patients with a specific molecular alteration could be the basis
for the design of a phase II-III trial. This transition from a phase I-II trial to a phase III trial
will save time and resources thus speeding the development process. A phase I trial is
designed to confirm a safe dose and an extension cohort at the recommended phase II dose
can evaluate efficacy in a specific subpopulation of prescreened patients - as performed in the
development of vemurafenib in b-RAF mutated metastatic melanoma and crizotinib in
anaplastic lymphoma kinase-mutated non-small cell lung cancer (36, 37). The design is not
necessarily based on the selection of patients by tumor subtypes and might be based on
molecular alterations. Different tumor types with a similar molecular phenotype could be
included in the phase I-II trial, and based on the clinical efficacy observed, a series of phase
II-III trials with adaptive design could be launched in specific tumor subtypes.
Patients with newly diagnosed metastatic cancer could be prescreened, and when they
progress on standard treatment, the molecular information could be used to randomize them
in an adaptive phase II-III trial with drug combinations (figure 2). The trial might evaluate
different arms that include a targeted agent alone or in combination with another modulator
of a defective molecular pathway, or with chemotherapy if there is preclinical evidence of a
therapeutically beneficial interaction. Different doses or schedules might also be explored in
different arms of the phase II part. A surrogate endpoint like PFS can be used in the phase II
trial but with an ambitious magnitude of benefit to select the best study arm for further
development of the phase III trial. Those arms with lack of efficacy or substantial toxicity
11
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
will be dropped but the arm with more activity and better safety profile can be expanded to a
phase III trial with increased accrual and with a more appropriate endpoint such as OS.
Adaptive designs also have limitations. There is no way to capture very rare toxicities or
those that appear later after the study has finished, and the logistics and interpretation of
results can be more complicated. Indeed, their implementation has been slower than
expected. Different reasons may explain this lack of endorsement by investigators, including
the more complicated logistics, concerns about security, or increased chance of erroneous
positive conclusions (see FDA guideline for adaptive designs). The incorporation of
biomarkers also adds cost and requires a process of validation. Phase III trials could also be
large, depending on the magnitude of the expected benefit. In addition, the final marketed
price of a drug will be selected to maximize the profit that the pharmaceutical company has
estimated for that drug, and has little or no relationship to the cost of drug development.
However, even with these limitations, phase III trials with adaptive designs can overcome
some of the problems associated with traditional phase III trials.
Conflict of Interest Statement: All authors of this manuscript declare no conflicts of
interest.
Acknowledgement: No funding sources to be declared.
12
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
References
1.
Pazdur R. Endpoints for assessing drug activity in clinical trials. Oncologist 2008;13
Suppl 2: 19-21.
2.
Sargent D. What constitutes reasonable evidence of efficacy and effectiveness to
guide oncology treatment decisions? Oncologist 2010;15 Suppl 1: 19-23.
3.
Tsimberidou AM, Braiteh F, Stewart DJ, Kurzrock R. Ultimate fate of oncology drugs
approved by the us food and drug administration without a randomized Trial. J Clin Oncol
2009;27: 6243-50.
4.
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
5.
Stewart DJ, Kurzrock R. Cancer: the road to Amiens. J Clin Oncol 2009;27: 328-33.
6.
Ocana A, Tannock IF. When are "positive" clinical trials in oncology truly positive? J
Natl Cancer Inst 2011;103: 16-20.
7.
Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, 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.
8.
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.
9.
Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati A, et al. Paclitaxelcarboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med
2006;355: 2542-50.
10.
Shepherd FA, Rodrigues Pereira J, Ciuleanu T, Tan EH, Hirsh V, Thongprasert S, et
al. Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med 2005;353: 12332.
11.
Joulain F, Van Cutsem E, Iqbal S, Hoyle M, Allegra CJ. Aflibercept versus placebo in
combination with FOLFIRI in previously treated metastatic colorectal cancer (mCRC): Mean
overall survival (OS) estimation from a phase III trial (VELOUR). J Clin Oncol 30, 2012
(suppl; abstr 3602)
12.
Ioannidis JP, Evans SJ, Gotzsche PC, O'Neill RT, Altman DG, Schulz K, et al. Better
reporting of harms in randomized trials: an extension of the CONSORT statement. Ann
Intern Med 2004;141: 781-8.
13.
Seruga B, Sterling L, Wang L, Tannock IF. Reporting of serious adverse drug
reactions of targeted anticancer agents in pivotal phase III clinical trials. J Clin Oncol
2011;29: 174-85.
13
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
14.
Tsang R, Colley L, Lynd LD. Inadequate statistical power to detect clinically
significant differences in adverse event rates in randomized controlled trials. J Clin
Epidemiol 2009;62: 609-16.
15.
Niraula S, Seruga B, Ocana A, Shao T, Goldstein R, Tannock IF, et al. The Price We
Pay for Progress: A Meta-Analysis of Harms of Newly Approved Anticancer Drugs. J Clin
Oncol 2012; 30:3012-9.
16.
Ocana A, Amir E, Yeung C, Seruga B, Tannock IF. How valid are claims for synergy
in published clinical studies? Ann Oncol 2012; 23:2161-6.
17.
Wu L, Birle DC, Tannock IF. Effects of the mammalian target of rapamycin inhibitor
CCI-779 used alone or with chemotherapy on human prostate cancer cells and xenografts.
Cancer Res 2005;65: 2825-31.
18.
Kim JJ, Tannock IF. Repopulation of cancer cells during therapy: an important cause
of treatment failure. Nat Rev Cancer 2005;5: 516-25.
19.
http://www.fda.gov/scienceresearch/specialtopics/criticalpathinitiative/default.htm.
20.
Baselga J, Segalla JG, Roche H, Del Giglio A, Pinczowski H, Ciruelos EM, et al.
Sorafenib in Combination With Capecitabine: An Oral Regimen for Patients With HER2Negative Locally Advanced or Metastatic Breast Cancer. J Clin Oncol 2012;30: 1484-91.
21.
Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, 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.
22.
Motzer RJ, Hutson TE, Olsen MR, Hudes GR, Burke JM, Edenfield WJ, et al.
Randomized phase II trial of sunitinib on an intermittent versus continuous dosing schedule
as first-line therapy for advanced renal cell carcinoma. J Clin Oncol 2012;30: 1371-7.
23.
Sullivan R, Peppercorn J, Sikora K, Zalcberg J, Meropol NJ, Amir E, et al. Delivering
affordable cancer care in high-income countries. Lancet Oncol 2011;12: 933-80.
24.
Rascati KL. The $64,000 question--what is a quality-adjusted life-year worth?
Clinical therapeutics 2006;28: 1042-3.
25.
Earle CC, Chapman RH, Baker CS, Bell CM, Stone PW, Sandberg EA, et al.
Systematic overview of cost-utility assessments in oncology. J Clin Oncol 2000;18: 3302-17.
26.
Shih YC, Halpern MT. Economic evaluations of medical care interventions for cancer
patients: how, why, and what does it mean? CA Cancer J Clin 2008;58: 231-44.
27.http://www.nice.org.uk/newsroom/features/measuringeffectivenessandcosteffectivenessthe
qaly.jsp. Last update Nov 2011.
14
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
28.
207.
Berry DA. Adaptive clinical trials in oncology. Nat Rev Clin Oncol 20011;8: 199-
29.
Korn EL, Freidlin B. Outcome-adaptive randomization: is it useful? J Clin Oncol
2011;29:771-776.
30.
Adaptive Design Clinical Trials for Drugs and Biologics, Draft Guidance for Industry.
February2010.http://fda.gov/downloads/DrugsGuidanceComplianceRegulatoryInformation/G
uidances/UCM201790.pdf
31.
James N, Sydes M, Mason M, Clarke NW, Amos C, Anderson J et al. Celecoxib plus
hormone therapy versus hormone therapy alone for hormone-sensitive prostate cancer: First
results from STAMPEDE (MRC PR08, CRUK/06/019), a randomized controlled trial. J Clin
Oncol 30, 2012 (suppl 5; abstr 26)
32.
Amir E, Seruga B, Martinez-Lopez J, Kwong R, Pandiella A, Tannock IF, et al.
Oncogenic targets, magnitude of benefit, and market pricing of antineoplastic drugs. J Clin
Oncol 2011;29: 2543-9.
33.
Berthold DR, Gulamhusein A, Jackson JI, Tannock IF. The transition from phase II to
phase III studies. J Clin Oncol 2009;27: 1150-1.
34.
<http://wwwclinicaltrialsgov/ct2/show/NCT01042379?term=I-SPY2&rank=1>.
35.
Baselga J, Cortes J, Kim SB, Im SA, Hegg R, Im YH, et al. Pertuzumab plus
trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med;366: 109-19.
36.
Kwak EL, Camidge DR, Clark J, Shapiro GI, Maki RG, Ratain MJ. Clinical activity
observed in a phase I dose escalation trial of an oral c-met and ALK inhibitor, PF-02341066.
J Clin Oncol 27:15s, 2009 (suppl; abstr 3509)
37.
Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, Sosman JA, et al.
Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med 2012;363:
809-19.
38.
Albain KS, Nag SM, Calderillo-Ruiz G, Jordaan JP, Llombart AC, Pluzanska A, et al.
Gemcitabine plus Paclitaxel versus Paclitaxel monotherapy in patients with metastatic breast
cancer and prior anthracycline treatment. J Clin Oncol 2008;26: 3950-7.
39.
Gradishar WJ, Tjulandin S, Davidson N, Shaw H, Desai N, Bhar P, et al. Phase III
trial of nanoparticle albumin-bound paclitaxel compared with polyethylated castor oil-based
paclitaxel in women with breast cancer. J Clin Oncol 2005;23: 7794-803.
40.
Sparano JA, Vrdoljak E, Rixe O, Xu B, Manikhas A, Medina C, et al. Randomized
phase III trial of ixabepilone plus capecitabine versus capecitabine in patients with metastatic
breast cancer previously treated with an anthracycline and a taxane. J Clin Oncol 2010;28:
3256-63.
15
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
41.
O'Shaughnessy J, Miles D, Vukelja S, Moiseyenko V, Ayoub JP, Cervantes G, et al.
Superior survival with capecitabine plus docetaxel combination therapy in anthracyclinepretreated patients with advanced breast cancer: phase III trial results. J Clin Oncol 2002;20:
2812-23.
42.
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: 5538-46.
43.
Van Cutsem E, Twelves C, Cassidy J, Allman D, Bajetta E, Boyer M, et al. Oral
capecitabine compared with intravenous fluorouracil plus leucovorin in patients with
metastatic colorectal cancer: results of a large phase III study. J Clin Oncol 2001;19: 4097106.
44.
Goldberg RM, Sargent DJ, Morton RF, Fuchs CS, Ramanathan RK, Williamson SK et
al. A randomized controlled trial of fluorouracil plus leucovorin, irinotecan, and oxaliplatin
combinations in patients with previously untreated metastatic colorectal cancer. J Clin Oncol
2004;22: 23-30.
45.
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 multicentre randomised trial. Lancet 2000;355:
1041-7.
46.
Saltz LB, Cox JV, Blanke C, Rosen LS, Fehrenbacher L, Moore MJ, et al. Irinotecan
plus fluorouracil and leucovorin for metastatic colorectal cancer. Irinotecan Study Group. N
Engl J Med 2000;343: 905-14.
47.
Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, et al.
Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of
HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3,
open-label, randomised controlled trial. Lancet 2010;376: 687-97.
48.
Vermorken JB, Remenar E, Van Herpen C, Chung HC, Shen L, Sawaki A, et al.
Cisplatin, fluorouracil, and docetaxel in unresectable head and neck cancer. N Engl J Med
2007;357: 1695-704.
49.
Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in
advanced hepatocellular carcinoma. N Engl J Med 2008;359: 378-90.
50.
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: 2636-44.
51.
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
16
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
progressing after docetaxel treatment: a randomised open-label trial. Lancet 2010;376: 114754.
52.
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: 2271-81.
53.
Escudier B, Pluzanska A, Koralewski P, Ravaud A, Bracarda S, Szczylik C, et al.
Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a
randomised, double-blind phase III trial. Lancet 2007;370: 2103-11.
54.
Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S et al. Efficacy of
everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled
phase III trial. Lancet 2008;372: 449-56.
55.
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: 125-34.
56.
Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, et al.
Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 2007;356:
115-24.
57.
Baselga J. Phase I and II clinical trials of trastuzumab. Ann Oncol 2001;12 Suppl 1:
S49-55.
58.
Ranson M, Hammond LA, Ferry D, Kris M, Tullo A, Murray PI, et al. ZD1839, a
selective oral epidermal growth factor receptor-tyrosine kinase inhibitor, is well tolerated and
active in patients with solid, malignant tumors: results of a phase I trial. J Clin Oncol
2002;20: 2240-50.
59.
Hidalgo M, Siu LL, Nemunaitis J, Rizzo J, Hammond LA, Takimoto C, et al. Phase I
and pharmacologic study of OSI-774, an epidermal growth factor receptor tyrosine kinase
inhibitor, in patients with advanced solid malignancies. J Clin Oncol 2001;19: 3267-79.
60.
Baselga J, Pfister D, Cooper MR, Cohen R, Burtness B, Bos M, et al. Phase I studies
of anti-epidermal growth factor receptor chimeric antibody C225 alone and in combination
with cisplatin. J Clin Oncol 2000;18: 904-14.
61.
Jonker DJ, O'Callaghan CJ, Karapetis CS, Zalcberg JR, Tu D, Au HJ, et al.
Cetuximab for the treatment of colorectal cancer. N Engl J Med 2007;357: 2040-8.
62.
Cunningham D, Humblet Y, Siena S, Khayat D, Bleiberg H, Santoro A, et al.
Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic
colorectal cancer. N Engl J Med 2004;351: 337-45.
63.
Vermorken JB, Mesia R, Rivera F, Remenar E, Kawecki A, Rottey S, et al. Platinumbased chemotherapy plus cetuximab in head and neck cancer. N Engl J Med 2008;359: 111627.
17
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
64.
Gordon MS, Margolin K, Talpaz M, Sledge GW Jr, Holmgren E, Benjamin R, et al.
Phase I safety and pharmacokinetic study of recombinant human anti-vascular endothelial
growth factor in patients with advanced cancer. J Clin Oncol 2001;19: 843-50.
65.
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 2004;350: 2335-42.
66.
Giantonio BJ, Catalano PJ, Meropol NJ, O'Dwyer PJ, Mitchell EP, Alberts SR, et al.
Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for
previously treated metastatic colorectal cancer: results from the Eastern Cooperative
Oncology Group Study E3200. J Clin Oncol 2007;25: 1539-44.
67.
Strumberg D, Richly H, Hilger RA, Schleucher N, Korfee S, Tewes M, et al. Phase I
clinical and pharmacokinetic study of the Novel Raf kinase and vascular endothelial growth
factor receptor inhibitor BAY 43-9006 in patients with advanced refractory solid tumors. J
Clin Oncol 2005;23: 965-72.
68.
Faivre S, Delbaldo C, Vera K, Robert C, Lozahic S, Lassau N, et al. Safety,
pharmacokinetic, and antitumor activity of SU11248, a novel oral multitarget tyrosine kinase
inhibitor, in patients with cancer. J Clin Oncol 2006;24: 25-35.
69.
Demetri GD, van Oosterom AT, Garrett CR, Blackstein ME, Shah MH, Verweij J, et
al. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour
after failure of imatinib: a randomised controlled trial. Lancet 2006;368: 1329-38.
70.
Weiner LM, Belldegrun AS, Crawford J, Tolcher AW, Lockbaum P, Arends RH et al.
Dose and schedule study of panitumumab monotherapy in patients with advanced solid
malignancies. Clin Cancer Res 2008;14: 502-8.
71.
Van Cutsem E, Peeters M, Siena S, Humblet Y, Hendlisz A, Neyns B, et al. Openlabel 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: 1658-64.
72.
Raymond E, Alexandre J, Faivre S, Vera K, Materman E, Boni J, et al. Safety and
pharmacokinetics of escalated doses of weekly intravenous infusion of CCI-779, a novel
mTOR inhibitor, in patients with cancer. J Clin Oncol 2004;22: 2336-47.
73.
O'Donnell A, Faivre S, Burris HA, 3rd, Rea D, Papadimitrakopoulou V, Shand N, et
al. Phase I pharmacokinetic and pharmacodynamic study of the oral mammalian target of
rapamycin inhibitor everolimus in patients with advanced solid tumors. J Clin Oncol
2008;26: 1588-95.
74.
Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, et al.
Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J
Med 2011;364: 2507-16.
18
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
75.
Kwak EL, Bang YJ, Camidge DR, Shaw AT, Solomon B, Maki RG, et al. Anaplastic
lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med 2010;363: 1693703.
19
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Table 1. RCTs used for the approval of agents from 2000 to 2011 that reported both OS
and PFS (or TTP, indicated by *) if this is given instead of PFS).
Tumor Type
Study design
HR: PFS/OS
0.70/0.78
CRC (43)
Gemcitabine+Paclitaxel vs
Paclitaxel*
Albumin-bound paclitaxel vs
Paclitaxel*
Ixabepilone+Capecitabine vs
Capecitabine
Capecitabine+Docetaxel vs
Docetaxel*
Letrozole + Lapatinib
vs Letrozole + Placebo
Capecitabine vs 5FU-LV*
CRC (44)
IFL vs FOLFOX vs IROX*
0.74/0.66
CRC (45)
IFC vs FC
NR/0.59
CRC (46)
IFL vs FL vs I
0.64/0.78
Gastric
cancer (47)
HNC (48)
Capecitabine or FU + Cisplatin +/Trastuzumab
TPF vs PF
0.71/0.74
0.72/0.73
HCC (49)
Sorafenib vs Placebo
1.08/0.69
Mesothe
lioma (50)
NSCLC (10)
Pemetrexed+Cisplatin vs Cisplatin*
0.68/0.77
Erlotinib vs Placebo
0.61/0.70
NSCLC (9)
Paclitaxel –Carboplatin+/Bevacizumab
Gemcitabine + Erlotinib vs
Gemcitabine + Placebo
Mitoxantrone + pred vs
Cabazitaxel+pred
Temsirolimus vs Interferon alfa vs
combination
0.66/0.79
Breast (38)
Breast (39)
Breast (40)
Breast (41)
Breast (42)
Pancreas (7)
Prostate (51)
Renal (52)
0.73/0.75
0.79/0.90
0.65/0.77
0.84
0.71/0.74
0.95
NR/0.92
0.77/0.82
0.74/0.70
NR/0.73
median PFS
(mos)
exp/control
difference
6.1/4.0
2.1
5.8/4.2
1.6
6.2/4.2
2.0
6.1/4.2
1.9
8.2/3.0
5.2
5.2-4.7
0.5
8.7/6.9
1.8
6.7/4.4
2.3
7.0/4.3
2.7
6.7/5.5
1.2
11/8.2
2.8
4.9/4.1
0.8
5.7/3.9
1.8
2.2/1.8
0.4
6.2/4.5
1.7
3.8/3.6
0.2
2.8/1.4
1.4
3.8/1.9
1.9
median OS (mos)
exp/control
difference
18.6/15.8
2.8
16.3/13.9
2.4
16.4/15.6
0.8
14.5/11.5
3.0
33.3/32.3
1.0
13.2/12.1
1.1
19.5/15.0
4.5
17.4/14.1
3.3
14.8/12.6
2.2
13.8/11.1
2.7
18.8/14.5
4.3
10.7/7.9
2.8
12.1/9.3
2.8
6.7/4.7
2.0
12.3/10.3
2.0
6.2/5.9
0.3
15.1/12.7
2.4
10.9/7.3
3.6
Ratio of
median
differences in
OS/PFS
1.33
1.50
0.40
1.58
0.19
2.20
2.50
1.43
0.81
2.25
1.53
3.50
1.55
5.00
1.18
1.50
1.71
1.89
Abbreviations: OS: overall survival; PFS: progression free survival; mos: months; NR: not recorded; CRC:
colorectal Cancer; HNC: Head and Neck Cancer; NSCLC: Non-small Cell Lung Cancer; *TTP: time to
treatment progression used instead of PFS; FU: fluorouracil; LV: leucovorin; I: irinotecan; IROX: irinotecan
and oxaliplatin; TPF: taxotere, platinum compound, fluorouracil; PF: platinum compound, fluorouracil.
20
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
2. Total cost of control and experimental arms, and estimated incremental cost per lifeyear gained for OS (or PFS if cross-over or if OS was not given), for new agents approved
for the treatment of renal cell carcinoma
Targeted agent
Design
Total
price:
control
arm (US$)
Total price:
experimental
arm (US$)
Primary
Endpoint
(PE)
Δ in median
OS/PFS
(months)
Incremental
cost per lifeyear gained
(US$)
Bevacizumab
(Bev)(53)
Everolimus (54)
Bev + IFN-α vs
IFN-α + Placebo
Everolimus vs
Placebo
31,260
157,220
PFS
314,900
Placebo
27,730
PFS
Not
reported/4.8
Not
reached/2.1
Sorafenib (55)
Sorafenib vs
Placebo
Sunitinib vs IFNα
Temsirolimus vs
IFN-α
Placebo
34,700
OS
3.4/2.7
122,500
7,720
52,260
PFS
89,000
5,870
21,440
OS
Not
reached/6.0
3.6/1.9
Sunitinib (56)
Temsirolimus
(52)
158,450
51,900
21
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Table 3. Evaluation of biomarkers for approved drugs from 2000 to 2011, from phase I
to phase III studies
Biomarker
included in
phase II-III
trials
Yes
No
Approved
indication
Phase III
trial for
approval
HER2
None
Selection of
patients based
on biomarker
in phase I
Yes
No
Breast
Lung
Slamon (21)
None
No
No
Lung
Shepherd (10)
EGFR for
CP02-9502 and
CP02-9401
Yes
No
Colon
Jonker (61)
Cunningham
(62)
Targeted agent
Publicati
on Year
Phase I
Target
Tumor type
Biomarker
Trastuzumab (57)
Gefitinib (58)
1999
2002
HER2
EGFR
Erlotinib (59)
2001
EGFR
Breast
Solid express
EGFR
Solid tumors
Cetuximab (60)
2002
EGFR
Solid tumors
with
EGFR overexpression
Head and
Neck cancer
Bevacizumab (64)
2001
VEGF
Solid tumors
None
No
No
Colon
NSCLC
Vermorken
(63)
Hurwitz (65)
Giantonio
(66)
Sandler (9)
RCC
Sorafenib (67)
2005
Sunitinib (68)
2006
Multi TKI
Multi TKI
Solid tumors
Solid tumors
pERK in PBL
VEGF, VGFR2
Panitumumab
(70)
Temsirolimus
(72)
2008
EGFR
Solid tumors
None
2004
mTOR
Solid tumors
None
Everolimus (73)
2008
mTOR
Solid tumors
pS6 in PBMC
Vemurafenib (37)
2010
B-RAF
Solid tumors
Erk1/2
Crizotinib (36)
2009
ALK
Solid tumors
No
(Signs of
activity in HCC
and RCC)
No
(Signs of
activity in RCC,
neuro-endocrine
and GIST)
None
No
(Signs of
activity in breast
cancer and
RCC)
No
(Signs of
activity in RCC)
No
(Signs of
activity in
melanoma)
No
(Signs of
activity in
sarcoma and
NSCLC)
HCC
Escudier (53)
Llovet (49)
RCC
Escudier (55)
RCC
Motzer (56)
GIST
Demetri (69)
No
Colon
No
RCC
Van Cutsem
(71)
Hudes (52)
No
RCC
Motzer (54)
Yes
Melanoma
Chapman (74)
Yes
NSCLC
Kwak (75)
No
No
Abbreviations: TKI: tyrosine kinase inhibitor; RCC: renal cell carcinoma; EGFR: epidermal growth factor
receptor; PBMC: peripheral blood monoclonal cells; RCC: renal cell carcinoma; NSCLC: non-small cell lung
cancer; GIST: gastrointestinal stromal tumors; PBL: peripheral blood lymphocytes; VEGFR: vascular
endothelial growth factor receptor; HCC: hepatocelular carcinoma.
22
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Figure 1. Forest plot showing meta-analyses of hazard ratios for progression-free
survival based on mechanism of action of targeted agents. Reproduced from “Oncogenic
Targets, Magnitude of Benefit, and Market Pricing of Antineoplastic Drugs” by Amir E, et al.
J Clin Oncol. 2011 Jun 20;29(18):2543-9, under permission of the American Society of
Clinical Oncology (number: 3173531452218)
Figure 2. Schematic representation of phase II- III adaptive designs using prescreened
populations.
23
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Figure 1:
Study or subgroup
Hazard ratio, 99% CI
Agents directed against a specific molecular target
Amado 2008
Demetri 2006
Geyer 2006
Karapetis 2008
Maemondo 2010
Mok 2009
Slamon 2001
Subtotal
3.0%
2.6%
2.5%
2.9%
2.9%
2.8%
3.3%
20.0%
0.45 [0.34, 0.59]
0.33 [0.23, 0.47]
0.49 [0.34, 0.71]
0.40 [0.30, 0.54]
0.30 [0.22, 0.41]
0.48 [0.35, 0.66]
0.51 [0.41, 0.63]
0.42 [0.36, 0.49]
Test for overall effect: Z = 11.00 (P<0.001)
Less specific biological targeted agents
Bonner 2006
3.0%
0.68 [0.52, 0.89]
Escudier 2007
3.1%
0.44 [0.34, 0.56]
Escudier 2007
3.4%
0.63 [0.52, 0.77]
Glantonio 2007
3.2%
0.61 [0.49, 0.77]
Hurwitz 2004
2.8%
0.54 [0.39, 0.74]
Jonker 2007
3.5%
0.68 [0.58, 0.80]
Llovet 2008
3.2%
0.58 [0.46, 0.73]
Miller 2007
3.5%
0.60 [0.51, 0.70]
Moore 2007
3.4%
0.77 [0.64, 0.93]
Motzer 2007
3.1%
0.42 [0.33, 0.54]
Motzer 2008
3.0%
0.31 [0.24, 0.41]
Sandler 2006
3.5%
0.66 [0.56, 0.77]
Shepherd 2005
3.4%
0.61 [0.51, 0.74]
Van Cutsem 2007
3.4%
0.54 [0.44, 0.66]
Subtotal
45.6% 0.57 [0.51, 0.64]
Test for overall effect: Z = 10.10 (P<0.001)
Chemotherapies
Albain 2008
3.5%
0.70 [0.59, 0.84]
Culeanu 2009
3.4%
0.50 [0.41, 0.61]
Goldberg 2004
3.4%
0.74 [0.61, 0.90]
Gradishar 2005
3.5%
0.75 [0.64, 0.89]
Hanna 2004
3.4%
0.97 [0.80, 1.18]
O’Shaughnessy 2002
3.4%
0.65 [0.53, 0.79]
Scagliotti 2008
3.6%
1.04 [0.92, 1.18]
Sparano 2010
3.6%
0.79 [0.69, 0.91]
Thomas 2007
3.5%
0.75 [0.64, 0.88]
Vogelzang 2003
3.2%
0.68 [0.54, 0.86]
Subtotal
34.4% 0.75 [0.66, 0.85]
Test for overall effect: Z = 4.39 (P<0.001)
Total (95% CI)
100.0% 0.59 [0.53, 0.65]
0.5 0.7 1 1.5 2
Favors experimental Favors control
Test for overall effect: Z = 10.46 (P<0.001)
Test for subgroup differences: ChF = 31.43, df = 2 (P<0.001)
CCR Reviews
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Standard treatment:
Chemotherapy
Biomarker +
Progression
Arm X: Standard
treatment
Experimental treatment:
Biological agent
Experimental treatment:
8_ebe]_YWbW][djiY^[Zkb[7
+ Chemotherapy
Experimental treatment:
8_ebe]_YWbW][djiY^[Zkb[8
+ Chemotherapy
Prescreened
population
Arm Y: Best
experimental treatment
Phase II
randomized part
Phase III part
š ;Whboijeff_d]hkb[i\eh
ineffective or toxic arms
š Ikhhe]Wj[[dZfe_dji
like PFS
š ?Z[dj_\ocW]d_jkZ[e\
X[d[\_j\ehf^Wi[???Z[i_]d
š I[b[YjX[ijZei[_\
_dYbkZ[Z_dWdoWhc
š 8[ijWhc_Z[dj_\_[Z_d
f^Wi[??fWhj
š 7ffhefh_Wj[[dZfe_djb_a[EI
š 7h[b[lWdjcW]d_jkZ[e\X[d[\_j
š 7Z[gkWj[iWcfb[i_p[je
detect clinical differences
WdZ[lWbkWj[jen_Y_jofhe\_b[
(rare toxicities)
© 2013 American Association for Cancer Research
CCR Reviews
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.
Figure 2:
Author Manuscript Published OnlineFirst on July 23, 2013; DOI: 10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
Phase III Trials of Targeted Anti-cancer Therapies: redesigning
the concept
Alberto Ocana, Eitan Amir, Francisco Vera-Badillo, et al.
Clin Cancer Res Published OnlineFirst July 23, 2013.
Updated version
Author
Manuscript
E-mail alerts
Reprints and
Subscriptions
Permissions
Access the most recent version of this article at:
doi:10.1158/1078-0432.CCR-13-1222
Author manuscripts have been peer reviewed and accepted for publication but have not yet been
edited.
Sign up to receive free email-alerts related to this article or journal.
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Department at [email protected].
To request permission to re-use all or part of this article, contact the AACR Publications
Department at [email protected].
Downloaded from clincancerres.aacrjournals.org on April 28, 2017. © 2013 American Association for Cancer
Research.