throbber
V O L U M E 2 6 䡠 N U M B E R 8 䡠 M A R C H 1 0 2 0 0 8
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`R E V I E W A R T I C L E
`
`Review of Phase II Trial Designs Used in Studies of
`Molecular Targeted Agents: Outcomes and Predictors of
`Success in Phase III
`Robert H. El-Maraghi and Elizabeth A. Eisenhauer
`
`A
`
`B
`
`S
`
`T
`
`R
`
`A
`
`C
`
`T
`
`Purpose
`Because the appropriate design and end points for phase II evaluation of targeted anticancer
`agents are unclear, we undertook a review of recent reports of phase II trials of targeted agents
`to determine the types of designs used, the planned end points, the outcomes, and the
`relationship between trial outcomes and regulatory approval.
`Methods
`We retrieved reports of single-agent phase II trials in six solid tumors for 19 targeted drugs. For
`each, we abstracted data regarding planned design and actual results. Response rates were
`examined for any relationship to eventual success of the agents, as determined by US Food and
`Drug Administration approval for at least one indication.
`Results
`Eighty-nine trials were identified. Objective response was the primary or coprimary end point in
`the majority of trials (61 of 89 trials). Fourteen reports were of randomized studies generally
`evaluating different doses of agents, not as controlled experiments. Enrichment for target expression
`was uncommon. Objective responses were seen in 38 trials; in 19 trials, response rates were more
`than 10%, and in eight, they were more than 20%. Agents with high response rates tended to have
`high nonprogression rates; renal cell carcinoma was the exception to this. Higher overall response
`rates were predictive of regulatory approval in the tumor types reviewed (P ⫽ .005).
`Conclusion
`In practice, phase II design for targeted agents is similar to that for cytotoxics. Objective response
`seems to be a useful end point for screening new targeted agents because, in our review, its
`observation predicted for eventual success. Improvements in design are recommended, as is
`more frequent inclusion of biological questions as part of phase II trials.
`
`J Clin Oncol 26:1346-1354. © 2008 by American Society of Clinical Oncology
`
`INTRODUCTION
`
`Recently, there has been a veritable explosion of
`knowledge with respect to the molecular biology of
`malignancy. This has led to the identification of po-
`tential new targets for cancer therapy and, subse-
`quently, to the rational design of agents created to
`affect those targets in a clinically meaningful way.
`However, as a growing number of agents targeting
`molecular pathways are tested in the clinic, there has
`been increasing pressure to rethink the standard
`drug development paradigm, specifically early trial
`design, to ensure that these promising new drugs are
`appropriately evaluated.
`The ultimate goal of drug development in on-
`cology is to identify new agents that provide a mean-
`ingful clinical benefit for patients, with the gold
`standard being the prolongation of patient survival.
`
`This is both a time- and resource-intensive process,
`with an estimated monetary cost to bring new drugs
`to market of $800 million (or more)1,2 with equally
`important, although perhaps less definable, costs to
`participating clinical subjects. With this in mind, it is
`understandable that there is pressure to optimize
`early clinical trial design so as to minimize the re-
`sources expended on drugs that are likely to fail in
`later development.
`The primary objective of phase II trials, regard-
`less of the nature of the compound, is to screen for
`preliminary evidence of efficacy in a given tumor
`type. For cytotoxic agents, the standard approach
`has been to enroll small numbers of patients in a
`nonrandomized design, often of two or more ac-
`crual stages, and use objective tumor regression as-
`sessed by standard criteria3,4 as the end point to
`identify drugs with potential efficacy. Retrospective
`
`From the Royal Victoria Hospital, Barrie;
`and National Cancer Institute of Canada
`Clinical Trials Group, Queen’s Univer-
`sity, Kingston, Ontario, Canada.
`
`Submitted July 18, 2007; accepted
`November 1, 2007; published online
`ahead of print at www.jco.org on
`February 19, 2008.
`
`The National Cancer Institute of Canada
`(NCIC) Clinical Trials Group (CTG) is
`supported by a core grant from the
`National Cancer Institute of Canada
`with funds received from the Canadian
`Cancer Society. R.H.E.-M. was a Drug
`Development Fellow at NCIC CTG from
`2005 to 2006 whose salary was
`supported by a Transdisciplinary Train-
`ing Program Grant from the Canadian
`Institutes of Health Research as well as
`a Drug Development Fellowship grant
`from AstraZeneca.
`
`Presented in part at the 42nd Annual
`Meeting of the American Society of
`Clinical Oncology, June 2-6, 2006,
`Atlanta, GA; and the National Cancer
`Research Institute Cancer Conference,
`October 8-11, 2006, Birmingham,
`United Kingdom.
`
`Authors’ disclosures of potential con-
`flicts of interest and author contribu-
`tions are found at the end of this
`article.
`
`Corresponding author: Elizabeth Eisen-
`hauer, MD, FRCPC, National Cancer
`Institute of Canada Clinical Trials Group,
`Queen’s University, 10 Stuart St, King-
`ston, Ontario, Canada K7L 3N6; e-mail:
`eeisenhauer@ctg.queensu.ca.
`
`© 2008 by American Society of Clinical
`Oncology
`
`0732-183X/08/2608-1346/$20.00
`
`DOI: 10.1200/JCO.2007.13.5913
`
`1346
`
`MYLAN - EXHIBIT 1023
`
`

`
`Phase II Designs Used for Targeted Therapy
`
`data support the use of the response end point for studies of cytotoxic
`agents5-7 and the efficiency of the nonrandomized design. The inher-
`ent differences in the mechanism of action between traditional cyto-
`toxic drugs and molecularly targeted agents, coupled with interest in
`increasing the reliability of phase II results in identifying truly active
`agents, have led to considerable discussion about the so-called tradi-
`tional approach to screening trials, particularly in regards to patient
`selection criteria, end points, and study design.
`With respect to patient selection, it is reasonable to expect that
`not all patients with a given tumor type will have similar levels of target
`protein activity or expression, and thus, efficacy of the targeted agent
`may vary according to which subpopulation is evaluated. Enriching
`the population to maximize possible activity could be achieved
`through restriction of study entry to those with a specific tumor
`histology or those whose tumors (over)express a molecular target.
`However, at the time of early clinical studies, such predictors of activity
`are by definition hypothetical, unless the agent affects a target that has
`had validated predictors identified through earlier clinical trials of
`other drugs affecting the same target.
`The appropriate end point for phase II trials of targeted agents
`has also been debated.8-12 Because many of these agents may affect
`tumor cells by reducing proliferation, rather than by causing cell
`death, the impact on tumor growth may be stabilization of disease or
`minor tumor shrinkage. Thus, it is argued that focusing only on
`objective response could result in overlooking some agents that could
`improve survival by causing disease stabilization. Indeed, in lung can-
`cer, attainment of stable disease, in addition to responses, has been
`shown to contribute to improved survival.13
`The design of phase II trials and the choice of which end point is
`to be used are closely linked; the use of a nonrandomized design in the
`traditional phase II trial is reasonable because objective responses are
`unlikely to be due to natural disease processes, rendering a control
`group unnecessary. However, even if response is believed to be a
`reasonable end point for trials of particular agents, the sample size of a
`nonrandomized trial may need adjustment. This is necessary if the
`hypothesized response rate of interest for the targeted agent is lower
`than what might be considered standard for cytotoxic agents, as would
`be the case if the trial population is unselected for predictive markers.
`When objective response alone is not foreseen to be a useful end
`point, novel designs have been proposed for phase II screening trials.
`Included is a multinomial design in which decisions about early stop-
`ping and conclusions on activity are based not only on the number of
`responses seen, but also on the proportion of patients demonstrating
`early disease progression.14,15 Another approach of interest is the
`randomized discontinuation trial where patients are treated with a
`new agent for a specified period of time, after which those with stable
`disease are randomly assigned to continue or discontinue therapy.16,17
`The end point for this type of trial is either time to event (eg, progres-
`sion) or the proportion of patients progression free at a specific time
`point after random assignment. This design also provides information
`on the activity of the drug in terms of rates of response and progression
`at the end of the run-in phase. The randomized discontinuation de-
`sign has been promoted to be of particular help in screening cytostatic
`drugs, such as molecular targeted agents, by permitting early assess-
`ment of whether delays in progression are related to treatment or
`disease and whether they are of sufficient magnitude to suggest that
`the drug may be effective.
`
`The issues we have just highlighted illustrate the fact that numer-
`ous proposals regarding changes to traditional phase II trial design and
`end points have been made in recent years. However, there is little
`information on the impact of these discussions on the actual end
`points and designs used for the evaluation of novel targeted agents in
`phase II trials. In this article, we report the results of a review of phase
`II designs, end points, and outcomes of a series for targeted agents that
`have been studied in clinical trials in the last few years. In particular, we
`were interested in determining the planned design and end point(s) in
`these trials, what the observed results were, and, when possible,
`whether the results of phase II trials of individual agents were useful in
`predicting which agents achieve regulatory approval. Our interest in
`this last point speaks to the ultimate goal of phase II screening trials
`regardless of design: Is the output of the trials useful in identifying
`those agents likely to succeed in phase III trials?
`
`METHODS
`
`Agents
`For this review, we focused on the 31 targeted agents that were the basis of
`a review of phase I trial design published in 2004.18 These agents have distinct
`intracellular or extracellular targets in pathways and can be grouped into major
`classes on the basis of the chemistry of the agent (small molecule, antisense
`oligonucleotide, or antibody) as well as the expected molecular target (Table
`1).19-83 All of these agents completed phase I investigation long enough in the
`past that phase II trials, if undertaken, should now be reported.
`
`Tumor Types and Search Strategy
`We defined six common solid tumor types (breast, colorectal, lung,
`ovarian, prostate, and renal cell carcinomas) on which to focus. We performed
`a MEDLINE search for published articles of completed phase II single-agent
`studies for all of the 31 drugs limited to the six tumor types noted. Studies using
`the agents in combination and studies in progress were excluded from the
`review. The search terms included the name of the agent (including trade
`name, if applicable) and the molecular target, with the limit of clinical trials,
`phase II. A cutoff date of April 30, 2006 was used for retrieving publications.
`In addition to full publications, we also attempted to identify final reports
`of phase II studies published as abstracts that were not yet reported in full
`article form. Abstracts were identified using an electronic search of the pro-
`ceedings of the American Society of Clinical Oncology, the American Associ-
`ation for Cancer Research, and the San Antonio Breast Cancer Symposia
`meetings until the end of 2005. Identified abstracts were excluded if they
`indicated continuing patient accrual or incomplete efficacy results. Abstract
`data were verified against data presented at the actual scientific meeting,
`when possible.
`
`Definition of Trial Versus Report
`Each publication, whether abstract or full article, was considered a re-
`port. A small number of reports included more than one trial. Examples were
`reports in which multiple tumor types were included (each analyzed sepa-
`rately), multiple targeted agents were evaluated (in a noncomparative ran-
`domized design), or multiple doses of the same targeted agent were tested. To
`describe the outcomes of each active arm of targeted therapy appropriately, we
`considered a study with n prospective active treatment groups in which there
`was no intent to conduct a formal statistical comparison to be n trials within a
`single report. For those few reports where random assignment was to standard
`therapy or placebo, the study arms with no active targeted therapy or placebo
`were not considered as separate trials, and the reported outcome was that
`which was planned for the overall study.
`
`Data Abstraction
`All data were abstracted independently for each trial by both authors,
`with any discrepancies resolved by consensus. A summary of abstract data is
`found in Appendix Table A1 (online only). All planned primary and secondary
`
`www.jco.org
`
`1347
`
`

`
`El-Maraghi and Eisenhauer
`
`Table 1. Targeted Agents From 2004 Phase I Review Subjected to Search for Phase II Results
`
`Phase I Review Agents
`
`This Phase II Review
`
`Target Category
`
`Target
`
`Agent
`
`EGFR/HER-2 signaling pathways
`
`Farnesyltransferase
`
`MEK
`mTOR
`Raf kinase
`
`Cell surface receptors
`
`EGFR
`
`Angiogenesis
`
`HER-2
`c-kit
`VEGF
`VEGFR (plus other targets)
`
`Extracellular matrix
`
`Other
`Matrix metalloproteinase
`
`Other
`
`Total No.
`
`BCL-2
`PKC␣
`DNA methyltransferase
`
`BMS-214662
`R115777
`L778, 123
`SCH 66336
`CI-1040
`CCI-779 (temsirolimus)
`ISIS 5132
`BAY 43-9006 (sorafenib)
`ZD1839 (gefitinib)
`OSI-774 (erlotinib)
`C225 (cetuximab)
`MAb225
`EMD 72000
`EKB 569
`RG83852
`Trastuzumab
`STI-571 (imatinib)
`Bevacizumab
`ZD6474
`PTK787
`SU6668
`SU5416 (semaxanib)
`SU11248 (sunitinib)
`Endostatin
`BB-2516 (marimastat)
`BAY 12-9566 (tanomastat)
`COL-3
`BMS-275291
`G3139
`ISIS 3521 (aprinocarsen)
`MG98
`31†
`
`Drug
`Type
`
`SM
`SM
`SM
`SM
`SM
`SM
`AS
`SM
`SM
`SM
`AB
`AB
`AB
`SM
`AB
`AB
`SM
`AB
`SM
`SM
`SM
`SM
`SM
`Other
`SM
`SM
`SM
`SM
`AS
`AS
`AS
`
`No. of Single-Agent
`Phase II Reports
`Identified
`
`No. of Single-Agent
`Phase II Trials in
`Selected Tumors*
`Reviewed
`
`Reference
`No.
`
`5
`
`1
`1
`3
`4
`1
`14
`5
`3
`
`8
`7
`2
`1
`
`2
`2
`
`1
`
`1
`
`5
`1
`65‡
`
`6
`
`1
`3
`7
`5
`2
`17
`5
`3
`
`9
`9
`5
`2
`
`2
`2
`
`3
`
`2
`
`5
`1
`89
`
`19-23
`
`24
`25
`26-28
`29-32
`33
`34-47
`48-52
`53-55
`
`56-63
`64-70
`71,72
`73
`
`74,75
`76,77
`
`78
`
`79
`
`29,32,80-82
`83
`
`Abbreviations: EGFR, epidermal growth factor receptor; HER-2, human epidermal growth factor receptor 2; SM, small molecule; MEK, mitogen-activated
`protein-Erk kinase; mTOR, mammalian target of rapamycin; AS, antisense oligonucleotide; AB, antibody; VEGF, vascular endothelial growth factor; VEGFR, vascular
`endothelial growth factor receptor; PKC␣, protein kinase C alpha.
`*Selected tumors were breast, lung (small-cell and non–small-cell), prostate, colorectal, renal, and ovary.
`†Nineteen agents identified for this review.
`‡Actual total is 67, but two reports have been counted twice because they each included two agents and thus are found twice in this table (ISIS 5312 and ISIS 3521).
`
`end points (eg, response, change in target expression, progression-free sur-
`vival, either individually or in a multinomial combination) were abstracted, as
`were the actual outcome measures (eg, number of patients enrolled/eligible,
`number of patients with complete response, partial response, stable disease,
`progressive disease [PD], median time to progression, and overall survival).
`Data were not collected with respect to sex, age, or nature of the prior systemic
`treatment of patients.
`Calculations
`Once data abstraction was completed, several computations were under-
`taken. To determine the total response rate for a given trial, the total number of
`patients achieving either complete or partial response was divided by all eligi-
`ble patients. Trials without response reported or collected were classified as not
`reported for this outcome. Although we initially planned to report stable
`disease rate, because substantially differing duration requirements were used
`to define stable disease across the trials reviewed, we elected to calculate the
`nonprogression rate as a means of better standardizing the output of our
`review. To determine the percentage of patients with nonprogression, patients
`with PD as best response were subtracted from the number of eligible patients
`
`entered to give the total number of nonprogressors (non-PD). This figure was
`then divided by the total number of eligible patients. Although we recognized
`that assignment of PD as a best response was somewhat dependent on the
`timing of follow-up (usually between 6 and 12 weeks), this was less variable
`than the duration used to define stable disease (which ranged from a few weeks
`to ⬎ 6 months).
`Once the response rate and PD rate for each trial was calculated, addi-
`tional summary information was generated for presentation in tabular form.
`This included the calculation of overall rates of response (and nonprogres-
`sion), which were determined for each drug by grouping all patients in all trials
`of a given agent with either complete or partial response (or nonprogression)
`and then dividing by the total number of eligible patients.
`Response rates for trials and for drugs were categorized in the following
`range groupings: 0%, more than 0% to ⱕ 10%, more than 10% to ⱕ 20%, and
`more than 20%. Nonprogression rates were categorized as follows: ⱕ 30%,
`more than 30% to ⱕ 50%, more than 50% to ⱕ 65%, and more than 65%. The
`numbers of trials with response rates or non-PD rates in the ranges noted were
`displayed in table format according to a variety of groupings of the trial data.
`
`1348
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`

`
`Phase II Designs Used for Targeted Therapy
`
`This included tables representing the number of trials in the various response
`categories by disease type, by individual drugs, by drugs grouped by target, and
`by population enrichment.
`Drug Approval
`Information on whether the agents under evaluation had received accel-
`erated or full US Food and Drug Administration (FDA) approval for use in any
`of the six tumor types as of June 2007 was also identified. A table was then
`created that listed the number of drugs with overall response rates in the ranges
`described earlier versus the number of drugs in each grouping that had
`achieved regulatory approval. The relationship between the four response
`categories and the probability of FDA approval was assessed by the exact
`Cochran-Armitage linear trend test.
`Agents With No Phase II Trials
`There were 12 agents from the original list of 31 for which no phase II
`trials in any of the six tumor types were found. Every reasonable effort was
`made to determine the reasons for this (eg, drug stopped development,
`skipped phase II altogether, was evaluated in other diseases than those we
`focused on, or went into combination phase II immediately after phase I).
`However, not all agents could be traced.
`
`RESULTS
`
`Agents and Trials
`Of the 31 agents surveyed, reports on single-agent phase II eval-
`uation were retrieved for 19 in at least one of the prespecified tumor
`types (Table 1).19-83 Altogether, 65 reports were identified (53 articles
`and 12 abstracts). Several reports contained results of evaluation of
`more than one tumor type or involved several different agents or dose
`levels; thus, the final tally of trials was 89. These were spread across all
`six tumor types, with the largest numbers in breast (21 trials) and lung
`cancers (13 trials in non–small-cell lung cancer and nine trials in
`small-cell lung cancer), followed by renal (15 trials), prostate (14
`trials), and ovarian cancers (six trials; Appendix Table A2, on-
`line only).
`
`Trial Design
`In the majority of
`Randomized versus nonrandomized designs.
`reports (51 of 65 reports; 78%), the investigational agent was evaluated
`using a nonrandomized single-arm design (Table 2). Randomization
`
`Table 2. Trial Design and End Points
`
`Design and End Point
`
`Nonrandomized
`Randomized
`Comparator arms:
`Placebo/standard
`Other investigational drug
`Other dose of same agent
`Primary end point
`Objective response
`Multinomial (response and
`progressive disease)
`Proportion progression free
`Progression-free survival
`Other
`
`No. of Reports
`(N ⫽ 65)
`
`51
`14
`
`Trials
`(N ⫽ 89)
`
`No.
`
`62
`27
`
`3
`4
`20
`
`51
`10
`
`8
`8
`12
`
`%
`
`70
`30
`
`57
`11
`
`9
`9
`13
`
`was used in 14 (22%) of 65 reports. However, in only two reports was
`there random assignment to a placebo (this included the randomized
`discontinuation phase of the BAY 43-9006 renal cell carcinoma trial33
`and a randomized phase II study of gefitinib v placebo in prostate
`cancer45). One study in prostate cancer randomly assigned patients
`between an experimental agent and an active corticosteroid control
`arm.74 In two reports,29,32 patients were randomly assigned between
`two different investigational drugs; in both, the random assignment
`was between ISIS 3521 (aprinocarsen) and ISIS 5132 in a noncom-
`parative phase II design. Finally, in nine reports, the random assign-
`ment was between various dose levels of the same investigational drug.
`End points. The primary end point on which the trial design was
`based was most commonly objective response (51 trials; 57%; Table
`2). In addition, 10 studies used a multinomial end point incorporating
`both response and nonprogression. Thus, objective response was the
`primary or coprimary end point in 61 trials. Only 16 trials were
`designed with end points of progression-free survival or the propor-
`tion of patients progression free at a prespecified time point. Some of
`the remaining studies were in prostate cancer and used measures of
`prostate-specific antigen (PSA) change (eg, PSA response, change in
`slope of PSA increase) as the primary end point. In addition, in two
`reports, toxicity was the primary end point.
`Population enrichment. Efforts to enrich the population under
`evaluation by restricting entry to patients of a particular molecular or
`histologic tumor subtype were undertaken in 18 (20%) of 89 trials. As
`can be seen in Table 3, in 14 studies, enrichment was on the basis of a
`molecular marker assessed in tumor.
`Hypotheses used in design. For those trials in which objective
`response was the primary end point, we attempted to identify the
`hypotheses used to derive the sample size from the methods sections of
`articles because we postulated that response rates of interest might be
`lower for targeted agents than those that have been traditionally used
`for phase II trials of cytotoxic drugs. Unfortunately, of the 51 trials in
`which response was indicated to be the primary end point, only 27
`described the hypotheses that had led to design and the planned
`sample size. Of these, 20 based the sample size and, thus, stopping rules
`on response rates of interest of 20% or higher. In seven trials, response
`rates in the 10% to 15% range were targeted.
`Sample size. Not surprisingly, the mean planned sample size for
`the trials reviewed depended on the design. For trials in which objec-
`tive response was the end point, the mean maximum sample size
`planned was 56 patients (based on the data reported for 35 trials).
`When progression-free survival or non-PD was the end point used,
`the mean maximum sample size planned was 115, and for the multi-
`nomial design, the mean maximum sample size was 41.
`
`Trial Results: Response and Nonprogression
`Response rates. Seventy-six of 89 trials reported objective re-
`sponse outcomes. In total, 38 trials had overall response rates of 0%. In
`the other 38 trials, objective responses were seen; in 19 trials, response
`rates were more than 10%, and in eight trials, response rates were
`more than 20%.
`Appendix Table A3 (online only) shows the reported response
`rates categorized in the ranges shown for all trials sorted by agent.
`Appendix Table A4 (online only) displays the same data but sorted by
`tumor type. Trial response results for all agents affecting the same
`target are shown in Appendix Table A5 (for example, all epidermal
`growth factor receptor inhibitors trials are displayed in one line in the
`
`www.jco.org
`
`1349
`
`

`
`El-Maraghi and Eisenhauer
`
`Table 3. Population Enrichment
`
`Basis for Enrichment
`
`EGFR expression
`HER-2 expression
`Histology
`BAC
`Clear cell
`c-kit
`Other (SD at 12 weeks)
`Total
`
`No. of Trials
`(n ⫽ 18)
`
`3
`7
`
`1
`2
`4
`1
`18
`
`Agent*
`Erlotinib (n ⫽ 2); cetuximab (n ⫽ 1)
`Trastuzumab
`
`Tumor Type*
`Lung (n ⫽ 1); ovary (n ⫽ 1); CRC (n ⫽ 1)
`Breast (n ⫽ 5); lung (n ⫽ 1); ovary (n ⫽ 1)
`
`Erlotinib
`Bevacizumab
`Imatinib
`Sorafenib
`Trastuzumab (n ⫽ 7); imatinib (n ⫽ 4);
`erlotinib (n ⫽ 3); bevacizumab (n ⫽ 2);
`sorafenib (n ⫽ 1); cetuximab
`
`Lung
`Renal cell carcinoma
`Lung (n ⫽ 3); ovary (n ⫽ 1)
`Renal cell carcinoma
`
`Reference
`No.
`
`50,51,53
`56-59,61,62
`
`49
`72
`65,67,68
`33
`
`Abbreviations: EGFR, epidermal growth factor receptor; CRC, colorectal cancer; HER-2, human epidermal growth factor receptor 2; BAC, bronchioloalveolar
`carcinoma; SD, stable disease.
`*Numbers in parentheses represent No. of trials.
`
`table; table online only). Finally, Table 4 provides the overall response
`rate by agent, pooling results for all trials (across all tumor types).
`Nonprogression rates. Results for nonprogression rates were also
`tabulated, but not all are shown. Table 4 lists the non-PD rates by agent
`(pooling across all trials for each particular agent). As can be seen,
`non-PD rates were variable, but several agents (sorafenib, cetuximab,
`temsirolimus, trastuzumab, and gefitinib) had non-PD rates of 50%
`or more overall.
`Although in most tumor types the ranking of agents by response
`rates or non-PD rates was similar (data not shown), renal cell carci-
`noma trials seemed to display a different pattern. Appendix Table A6
`(online only) shows overall response rate and non-PD rates in renal
`cell carcinoma studies by agent. High non-PD rates were seen with
`four agents (sunitinib, sorafenib, temsirolimus, and imatinib), but
`
`only one of these, sunitinib, had a response rate that was more than
`20%; the remainder had observed response rates less than 10%.
`
`Response and Non-PD Rates by Disease Type and
`Regulatory Approval
`To identify whether there were tumor-related patterns in the
`response or non-PD results and regulatory approval (as of June
`2007), we examined overall rates of non-PD and response by agent
`in each tumor type in the trials reviewed, as shown in Appendix
`Tables A7 and A8 (online only). Numbers within each tumor type
`are too small to apply statistics, but it was observed that no agent
`with a 0% response rate in a given tumor type received approval in
`that tumor type. Similarly, no agent with non-PD rates less than
`30% in a given tumor type received approval in that tumor type.
`
`Table 4. Overall Response and Non-PD Rates by Agent
`
`No. of Trials
`
`Overall Response
`Rate (%)
`
`Total No. of Patients
`in Response Rate
`Denominator*
`
`Overall Non-PD
`Rate (%)
`
`Total No. of Patients
`in Non-PD Rate
`Denominator*
`
`2
`3
`5
`2
`3
`7
`3
`5
`5
`1
`5
`6
`1
`9
`2
`2
`9
`17
`2
`89
`
`4
`NR
`5.3
`0
`3
`8
`0
`0
`0
`0
`12
`5
`0
`0
`28
`5
`18
`10
`0
`
`202
`NA
`97
`80
`169
`190
`52
`87
`71
`15
`200
`208
`21
`112
`127
`45
`562
`698
`46
`
`75
`NR
`NR
`36
`60
`66
`12
`13
`27
`40
`39
`23
`14
`21
`0
`0
`53
`50
`NR
`
`202
`NA
`NA
`80
`58
`190
`52
`60
`71
`15
`150
`208
`21
`112
`63
`29
`420
`627
`NA
`
`Agent
`
`Sorafenib
`Marimastat
`Bevacizumab
`BMS-275291
`Cetuximab
`Temsirolimus
`CI-1040
`Aprinocarsen
`ISIS 5132
`MG98
`Erlotinib
`R1155777
`SCH 66336
`Imatinib
`Sunitinib
`Semaxanib
`Trastuzumab
`Gefitinib
`ZD6474
`Total
`
`Abbreviations: PD, progressive disease; NR, not reported in any trial; NA, not applicable.
`*Response and non-PD rate denominators may not match if some trials had one or the other not reported. Only trials with data reported were used to calculate rates.
`
`1350
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`

`
`Phase II Designs Used for Targeted Therapy
`
`What seems of interest is that, despite the suggestion that targeted
`drugs could not be screened by assessment of tumor shrinkage, the
`evidence suggests otherwise; of the 89 trials, 38 documented objective
`tumor responses, and 19 had response rates of more than 10% (and
`eight had response rates of ⬎ 20%). When examined by drug, of the 19
`drugs reviewed, 10 produced overall response rates (across all tumor
`types) ranging from 3% to 28%. Of these, seven have received full FDA
`approval for one or more indications. Furthermore, the data suggest a
`relationship between the level of the overall phase II response rate seen
`in the tumor types reviewed and the likelihood of achieving regula-
`tory approval.
`These results, although based on a relatively small number of
`targeted agents, are similar to a recent review of cytotoxic agents by
`Goffin et al.7 In that review, phase II outcomes of 46 cytotoxic drugs
`were compiled. The authors found a relationship between eventual
`regulatory approval and phase II response rate; drugs with overall
`response rates greater than 20% had a higher likelihood of approval
`than those with response rates of 10% to 20%, which, in turn, were
`more likely to receive approval than those with response rates of 0% or
`0% to 10%. The relationship seemed to hold true for breast, non–
`small-cell lung, ovary, and colorectal cancers but not for renal cell
`carcinoma or melanoma, although the numbers of randomized trials
`actually conducted in the latter two tumor types were fewer. In an
`accompanying editorial, Ratain11 argued that looking at the data as if
`the phase II response rates were diagnostic test results might be more
`appropriate. When he reanalyzed the data in this fashion, he identified
`that the negative predictive value of phase II results was quite high, but
`the positive predictive value was lower, being 0% in melanoma and
`renal cell carcinoma and from 33% to 75% for breast, ovarian, colo-
`rectal, and non–small-cell lung cancers when overall response rates
`were 20% or more. The problem with the approach of viewing the
`response rate outcome as if it were a diagnostic test is that it is guaran-
`teed to achieve high negative predictive values if no randomized trials
`are ever performed to assess survival impact when response rates are
`zero or low.
`Our review has limitations. First, not all targeted agents studied in
`the last decade were included. We identified phase II reports from the
`list of 31 targeted agents reviewed recently with respect to phase I
`design as a convenient approach to following up on the clinical devel-
`opment of those drugs. Second, we did not attempt to identify whether
`trials were conducted but never reported. Furthermore, we arbitrarily
`focused on six common solid tumor types. Thus, the overall single-
`agent response rates cited for each agent could be subject to some
`variation if data from unpublished studies or if trials in all tumor types
`had been included. In addition, our list of the original 31 agents was
`reduced to 19 because 12 drugs had no reported phase II single-agent
`studies in the tumor types that were of interest. Several of those agents
`were dropped from development after phase I, and others either
`remain in early phase II development or skipped phase II altogether to
`proceed into randomized combination studies.
`Nonetheless, there are interesting messages here. First, even rela-
`tively low rates of objective response may signal that an agent has
`potential for achieving regulatory approval on the basis of subsequent
`randomized data. It can be inferred from this observation that agents
`affecting targets that are meaningful in one or more cancer types
`should reasonably be expected to cause tumor shrinkage in at least
`some patients. Failing to see any evidence of response at all suggests
`that the drug is likely to fail in subsequent development. There were no
`
`Noteworthy was the observation that three of four agents in renal
`cell carcinoma with non-PD rates of more than 65% have been
`approved for renal cell carcinoma.
`
`Overall Response Rate and Regulatory Approval
`A number of the agents reviewed in this article had received FDA
`approval by June 2007. Figure 1 graphically displays the relationship
`between overall response rate for each agent (including all trials and all
`tumor types) and regulatory approval. Table 5 provides the same
`information clustering agents into the four response categories used
`earlier. Overall, seven of the 19 agents reviewed have received full FDA
`approval; one (gefitinib) received accelerated approval but failed to
`achieve full approval on the basis of randomized data. The P value of
`the exact Cochran-Armitage trend test for the relationship between
`response category into which agents fell and regulatory approval was
`P ⫽ .005 if the accelerated approval for gefitinib was not counted as
`FDA approved and was P ⬍ .0001 if the accelerated approval for
`gefi

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket