throbber
NIH Public Access
`Author Manuscript
`J Clin Oncol. Author manuscript; available in PMC 2014 May 05.
`
`Published in final edited form as:
`J Clin Oncol. 2008 March 1; 26(7): 1148–1159. doi:10.1200/JCO.2007.12.4487.
`
`Design and End Points of Clinical Trials for Patients With
`
`Progressive Prostate Cancer and Castrate Levels of
`
`Testosterone: Recommendations of the Prostate Cancer Clinical
`
`Trials Working Group
`
`Howard I. Scher, Susan Halabi, Ian Tannock, Michael Morris, Cora N. Sternberg, Michael A.
`
`Carducci, Mario A. Eisenberger, Celestia Higano, Glenn J. Bubley, Robert Dreicer, Daniel
`
`Petrylak, Philip Kantoff, Ethan Basch, William Kevin Kelly, William D. Figg, Eric J. Small,
`
`Tomasz M. Beer, George Wilding, Alison Martin, and Maha Hussain
`Sidney Kimmel Center for Prostate and Urologic Cancers, Memorial Sloan-Kettering Cancer
`Center, New York, NY; Duke University Medical Center, Durham, NC; Princess Margaret
`Hospital, Toronto, Ontario, Canada; Sam Camillo Forlanini Hospital, Rome, Italy; Beth Israel
`Deaconess Medical Center; Dana-Farber Cancer Center, Boston, MA; Sidney Kimmel
`Comprehensive Cancer Center at Johns Hopkins, Baltimore; National Cancer Institute, Bethesda,
`
`© 2008 by American Society of Clinical Oncology
`
`Corresponding author: Howard I. Scher, MD, Genitourinary Oncology Service, Department of Medicine, Sidney Kimmel Center for
`Prostate and Urologic Cancers, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY 10065;
`byczekb@mskcc.org.
`
`AUTHOR CONTRIBUTIONS
`Conception and design: Howard I. Scher, Susan Halabi, Ian Tannock, Michael Morris, Cora N. Sternberg, Michael A. Carducci,
`Mario A. Eisenberger, Celestia Higano, Glenn J. Bubley, Robert Dreicer, Daniel Petrylak, Philip Kantoff, Ethan Basch, William D.
`Figg, Eric J. Small, Alison Martin, Maha Hussain
`Provision of study materials or patients: Howard I. Scher
`Collection and assembly of data: Howard I. Scher, Mario A. Eisenberger, Celestia Higano, Tomasz M. Beer, Alison Martin
`Data analysis and interpretation: Howard I. Scher, Michael Morris, Michael A. Carducci, Mario A. Eisenberger, Glenn J. Bubley,
`Robert Dreicer, Philip Kantoff, William Kevin Kelly, William D. Figg, Tomasz M. Beer, George Wilding, Alison Martin, Susan
`Halabi, Maha Hussain
`Manuscript writing: Howard I. Scher, Susan Halabi, Ian Tannock, Michael Morris, Cora N. Sternberg, Michael A. Carducci, Mario
`A. Eisenberger, Celestia Higano, Glenn J. Bubley, Robert Dreicer, Ethan Basch, William Kevin Kelly, William D. Figg, Eric J. Small,
`Tomasz M. Beer, Alison Martin, Maha Hussain
`Final approval of manuscript: Howard I. Scher, Susan Halabi, Ian Tannock, Cora N. Sternberg, Michael A. Carducci, Mario A.
`Eisenberger, Celestia Higano, Glenn J. Bubley, Robert Dreicer, Daniel Petrylak, Philip Kantoff, Ethan Basch, William Kevin Kelly,
`William D. Figg, Eric J. Small, Tomasz M. Beer, George Wilding, Alison Martin, Maha Hussain
`
`AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
`Although all authors completed the disclosure declaration, the following author(s) indicated a financial or other interest that is relevant
`to the subject matter under consideration in this article. Certain relationships marked with a “U” are those for which no compensation
`was received; those relationships marked with a “C” were compensated. For a detailed description of the disclosure categories, or for
`more information about ASCO’s conflict of interest policy, please refer to the Author Disclosure Declaration and the Disclosures of
`Potential Conflicts of Interest section in Information for Contributors.
`Employment or Leadership Position: None Consultant or Advisory Role: Ian Tannock, sanofi-aventis (U), Algeta ASA (U), GPC
`Biotech (U); Michael A. Carducci, Abbott Laboratories (C), sanofi-aventis (U), Methylgene (C), Cougar Biotech (C); Mario A.
`Eisenberger, sanofi-aventis (C), GPC (C), Celgene (C); Robert Dreicer, Merck (C), sanofi-aventis (C), Bristol-Myers Squibb (C);
`Daniel Petrylak, Aventis (C), GPC Biotech (C), Abbott Laboratories (C); Eric J. Small, Cougar Biotechnology (C), Poniard
`Pharmaceuticals (C); Tomasz M. Beer, Novacea (C) Stock Ownership: Tomasz M. Beer, Novacea Honoraria: Michael A. Carducci,
`sanofi-aventis, Abbott Laboratories; Mario A. Eisenberger, sanofi-aventis, Ipsen, GPC; Robert Dreicer, Berlex; Daniel Petrylak,
`Aventis, Celegene, Abbott; William Kevin Kelly, sanofi-aventis, Genetech; Tomasz M. Beer, sanofi-aventis Research Funding: Ian
`Tannock, sanofi-aventis, Novacea; Mario A. Eisenberger, sanofi-aventis, Clegene, Cytogen; Robert Dreicer, sanofi-aventis, Eli Lilly,
`Millenium; Daniel Petrylak, Aventis, Celegene, GPC Biotech; William Kevin Kelly, sanofi-aventis, Genetech, Curagen; Eric J. Small,
`Dendreon, Novartis; Tomasz M. Beer, sanofi-aventis Expert Testimony: None Other Remuneration: None
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`MYLAN PHARMS. INC. EXHIBIT 1124 PAGE 1
`
`

`

`Scher et al.
`
`Page 2
`
`MD; University of Washington, Seattle, WA; Cleveland Clinic, Cleveland, OH; Columbia
`Presbyterian Medical Center, New York, NY; Yale Cancer Center, New Haven, CT; UCSF
`Comprehensive Cancer Center, San Francisco, CA; Oregon Health and Science Universeity,
`Portland, OR; University of Wisconsin Comprehensive Cancer Center, Madison, WI; and
`University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
`
`Abstract
`
`Purpose—To update eligibility and outcome measures in trials that evaluate systemic treatment
`
`for patients with progressive prostate cancer and castrate levels of testosterone.
`
`Methods—A committee of investigators experienced in conducting trials for prostate cancer
`
`defined new consensus criteria by reviewing previous criteria, Response Evaluation Criteria in
`
`Solid Tumors (RECIST), and emerging trial data.
`
`Results—The Prostate Cancer Clinical Trials Working Group (PCWG2) recommends a two-
`
`objective paradigm: (1) controlling, relieving, or eliminating disease manifestations that are
`
`present when treatment is initiated and (2) preventing or delaying disease manifestations expected
`
`to occur. Prostate cancers progressing despite castrate levels of testosterone are considered
`
`castration resistant and not hormone refractory. Eligibility is defined using standard disease
`
`assessments to authenticate disease progression, prior treatment, distinct clinical subtypes, and
`
`predictive models. Outcomes are reported independently for prostate-specific antigen (PSA),
`
`imaging, and clinical measures, avoiding grouped categorizations such as complete or partial
`
`response. In most trials, early changes in PSA and/or pain are not acted on without other evidence
`
`of disease progression, and treatment should be continued for at least 12 weeks to ensure adequate
`
`drug exposure. Bone scans are reported as “new lesions” or “no new lesions,” changes in soft-
`
`tissue disease assessed by RECIST, and pain using validated scales. Defining eligibility for
`
`prevent/delay end points requires attention to estimated event frequency and/or random
`
`assignment to a control group.
`
`Conclusion—PCWG2 recommends increasing emphasis on time-to-event end points (ie, failure
`
`to progress) as decision aids in proceeding from phase II to phase III trials. Recommendations will
`
`evolve as data are generated on the utility of intermediate end points to predict clinical benefit.
`
`INTRODUCTION
`
`Evaluating drugs to treat prostate cancer poses unique challenges. Measurable disease
`
`occurs infrequently, the natural history may be prolonged over decades, and because the
`
`treatment population is elderly, pursuing aggressive therapies may cause more harm than
`
`good. In 1999, the Prostate-Specific Antigen Working Group (PCWG1) addressed these
`challenges in their consensus recommendations for the conduct of clinical trials.1 They
`focused on trial development for patients with metastatic prostate cancer whose disease was
`
`progressing despite castrate levels of testosterone and defined eligibility and outcome
`
`measures based on clinically relevant end points, and proposed standards for the use of
`
`prostate-specific antigen (PSA). In 2000, a broader collective of cancer researchers
`
`introduced New Guidelines to Evaluate the Response to Treatment in Solid Tumors
`(Response Evaluation Criteria in Solid Tumors [RECIST]).2 This international initiative
`sought to standardize criteria to assess tumor response in trials for all solid tumors. Although
`
`J Clin Oncol. Author manuscript; available in PMC 2014 May 05.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`MYLAN PHARMS. INC. EXHIBIT 1124 PAGE 2
`
`

`

`Scher et al.
`
`Page 3
`
`RECIST served some cancer swell, its metrics did not capture some key characteristics of
`prostate cancer.3 For example, post-therapy changes in PSA, a routinely reported outcome in
`prostate cancer clinical trials and the primary focus of PCWG1, were not addressed by
`
`RECIST. In fact, none of the approved treatments for patients with prostate cancer would be
`
`available if trial outcomes were based solely on either the PCWG1 criteria or RECIST.
`
`Since these two initiatives were introduced, the biology and natural history of prostate
`
`cancer have become better understood, and diverse new therapies, including bone-targeted
`
`agents and signaling inhibitors, have become available for clinical testing. In 2004, the US
`
`Food and Drug Administration (FDA) challenged the prostate cancer clinical trials
`
`community to rework the eligibility and outcome measures from PCWG1 so they could be
`
`applied across the clinical spectrum of the disease. The subsequent process prompted the
`
`formation of the Prostate Cancer Clinical Trials Working Group (PCWG2), a collective of
`
`international investigators who developed this report through meetings and electronic
`
`communication.
`
`This article addresses clinical trials for patients with progressive prostate cancer despite
`
`castrate levels of testosterone and frames clinical trial questions for agents that act by
`
`diverse mechanisms. The consensus is that researchers should adopt a paradigm in which
`
`trial objectives are defined on the basis of controlling, relieving, or eliminating disease
`
`manifestations that are present when treatment is initiated, and/or of preventing or delaying
`
`disease manifestations expected to occur. This new paradigm expands the focus of prostate
`
`cancer clinical trials from traditional outcome measures such as early changes in PSA to
`
`time-to-event end points that capture the impact of treatment on important clinical
`
`manifestations and indicate when a drug should be stopped as the measure of antitumor
`
`effect. It also recommends standardized criteria for assessing patients. A goal of these
`
`recommendations is to ensure that a drug is not discontinued because of inappropriate
`
`outcome measures before it has had a chance to work.
`
`Although the intent of these guidelines is to maximize the ability of phase II trials to screen
`
`or select promising therapies, the eligibility and outcome measures have broad applicability
`
`and are relevant to the design and conduct of phase III trials. Incorporation of similar
`
`parameters into phase III trials assessing overall survival is encouraged to generate the
`
`databases that will allow validation or refinement of the intermediate end points proposed
`
`herein.
`
`I. CONCEPTUALIZING THE DISEASE
`
`Investigators need to adopt a common language to categorize the clinical spectrum of
`
`prostate cancer from diagnosis to metastasis. When PCWG1 was published, no common
`
`vocabulary was broadly accepted. PCWG2 categorizes the disease continuum of prostate
`
`cancer on the basis of whether metastases are detectable (clinically or by imaging) and
`
`whether the serum testosterone level is in the castrate range by a surgical orchiectomy or
`medical therapy (Fig 1).3,4 Each state on this continuum represents a scenario encountered
`routinely in clinical practice.
`
`J Clin Oncol. Author manuscript; available in PMC 2014 May 05.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`MYLAN PHARMS. INC. EXHIBIT 1124 PAGE 3
`
`

`

`Scher et al.
`
`Page 4
`
`The clinical-states model identifies patients with distinct prognoses who might benefit (or
`
`not) from specific therapeutic approaches. The rising PSA states (castrate and noncastrate)
`
`signify that no detectable metastatic disease was found in the past or is now present. The
`
`clinical metastases states (castrate and noncastrate) signify that disease was detectable at
`
`some point in the past, regardless of whether it is detectable now. Along this disease
`
`continuum, a patient can only advance. For example, a patient with radiographically evident
`
`bone metastases at diagnosis would be assigned to the clinical metastases–noncastrate
`
`disease state. If that patient is treated with androgen depletion, no longer has
`
`radiographically evident disease, and has a PSA level that is not rising, he remains
`
`categorized in the clinical metastases–noncastrate state.
`
`II. DEFINING THERAPEUTIC OBJECTIVES
`
`Since the publication of PCWG1 criteria, clinical investigators have used them to define the
`
`primary end points for phase II trials for prostate cancer patients with progressive,
`
`castration-resistant disease. These trials are designed to demonstrate whether the therapeutic
`
`effects observed justify further evaluation in large-scale phase III trials. Phase III trials
`
`characterize the risk/benefit profile of the treatment in relation to either a placebo or
`
`established standards, such as time to clinically relevant progression, survival, or quality of
`
`life. The clinical-states model offers investigators a framework to standardize phase II end
`
`points to appropriately inform phase III end points.
`
`PCWG2 distinguishes two types of phase II trial objectives: (1) those based on controlling,
`
`relieving, or eliminating disease manifestations that are present when treatment is initiated,
`
`and (2) those based on preventing or delaying future disease manifestations. Traditional
`
`measures of response reflect when a treatment is working; measures of progression indicate
`
`when a drug should be stopped. Because of the uncertainties associated with assessing
`
`response in bone and the controversy surrounding the clinical significance of post-therapy
`
`changes in PSA, PCWG2 recommends expanding the focus of phase II trials from measures
`
`of response to measures of progression. For most agents, a reliably determined, clinically
`
`relevant improvement in time to progression provides the most useful way to assess whether
`
`to proceed from a phase II to a phase III trial and may, if reproduced in a randomized,
`
`controlled trial, be evidence of clinical benefit from a regulatory perspective.
`
`The drug evaluation pathways for cytotoxic and noncytotoxic agents need to be developed
`
`separately. Cytotoxic drugs typically produce a decline in PSA and regression of target
`
`lesions, whereas agents that act to slow tumor growth, inhibit destruction of bone, or inhibit
`
`angiogenesis may not. For example, a bone-directed therapy may prevent disease-related
`
`complications in the skeleton without influencing the growth of soft-tissue disease.
`
`Depending on the agent and the study, PCWG2 recommends that the effects of cytotoxic
`
`drugs be assessed with both control/relieve/eliminate or prevent/delay end points, and
`
`noncytotoxic drugs with prevent/delay end points.
`
`Changes in existing manifestations of disease provide signals whether or not a treatment has
`
`produced an antitumor effect at an early stage, even though such changes may not
`
`necessarily signify clinical benefit. For example, a declining PSA level may be useful to
`
`J Clin Oncol. Author manuscript; available in PMC 2014 May 05.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`MYLAN PHARMS. INC. EXHIBIT 1124 PAGE 4
`
`

`

`Scher et al.
`
`Page 5
`
`screen for the activity of a cytotoxic agent, even though it does not mean that the patient will
`
`live longer. However, when designing trials with control, relieve, or eliminate end points for
`
`patients with symptoms, it is often difficult to distinguish whether a symptom is related to
`
`the cancer, prior treatment, comorbidities, or a combination of factors.
`
`Patients who lack discernible disease manifestations (eg, symptomatic bone pain), may be
`
`enrolled onto trials with prevent or delay end points that seek to prevent symptoms from
`
`occurring in the future. Manifestations that may occur in the future include growth at an
`
`existing site of disease, spread to additional sites, an increase in markers, new disease-
`
`related symptoms (eg, pain or other skeletal events), and death resulting from disease. The
`
`success of trials evaluating prevent or delay end points depends on the ability to define a
`
`patient cohort with a defined probability of developing the manifestations that the treatment
`
`is designed to prevent and in what time frame. Biases in interpreting the significance of
`
`time-to-event end points in phase II trials have been well described and support the case for
`randomized trial designs.5 Regardless of the end point, it is essential that the trial be
`designed in a way that does not allow a drug to be discontinued prematurely on the basis of
`
`criteria that do not reflect that the treatment was ineffective or failed to benefit the patient.
`
`III. ESTABLISHING ELIGIBILITY FOR ENROLLMENT
`
`After defining the primary end points of efficacy (either control/relieve/eliminate or prevent/
`
`delay), investigators can effectively set eligibility criteria. PCWG1 restricted enrollment in
`
`trials to patients with progressive disease despite castrate levels of testosterone, based on
`
`changes in PSA, measurable disease, and bone scan, while controlling for antiandrogen
`
`withdrawal responses to avoid the potential erroneous misattribution of response to a study
`
`agent. PCWG2 modifies these eligibility criteria by authenticating disease with standardized
`
`assessments, considering the prior treatment history in more detail, defining distinct clinical
`
`subtypes, and highlighting the importance of predictive models for future clinical events.
`
`The demonstration of a survival benefit in a phase III trial and a confirmatory trial7 led to
`the approval of docetaxel in 2004.6 Since then, clinical trials for patients with castrate
`metastatic disease are being designed in three contexts: before receiving treatment with
`
`docetaxel, with agents in combination with docetaxel to improve first-line outcomes, and as
`
`second-line treatment for patients with disease that has progressed despite docetaxel.
`
`Independent of the context, PCWG2 recommends defining therapeutic objectives in relation
`
`to the mechanism of action of the agent under study, documenting disease manifestations at
`
`the time treatment is started (Table 1), and serially evaluating patients post-treatment using
`
`standard assessments that relate to the objectives of the trial.
`
`Authenticating Disease Progression
`
`Authenticating disease progression is achieved by establishing standard pretreatment
`
`assessments and identifying standard criteria for disease progression for entry.
`
`Pretreatment assessments—PCWG1 did not define a standard pretreatment
`
`evaluation, so PCWG2 builds on the standards for base-line evaluations recommended by
`
`RECIST and provides guidelines for imaging and symptom assessment.
`
`J Clin Oncol. Author manuscript; available in PMC 2014 May 05.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`MYLAN PHARMS. INC. EXHIBIT 1124 PAGE 5
`
`

`

`Scher et al.
`
`Page 6
`
`Baseline evaluations—Baseline evaluations should be tailored both to target outcome
`
`measures and to contribute to the development of prognostic factors or other research
`
`questions. For the baseline evaluation, PCWG2 recommends documenting patient
`
`demographics, including age and performance status, clinical stage, PSA and Gleason score
`
`at the time of diagnosis, details and dates of the primary therapy (eg, pathologic stage and/or
`
`dose and type of radiation therapy as appropriate), and post-treatment PSA nadir. Details
`
`and dates of prior hormonal and nonhormonal therapies should be recorded, along with
`
`additional PSA measurements that can be used to estimate PSA doubling times (PSA-DTs).
`
`The presence or absence of disease in the primary site should also be documented.
`
`Imaging—PCWG2 pretreatment evaluations include imaging of the chest by plain
`
`radiograph or computed tomography (CT), a CT scan or magnetic resonance imaging (MRI)
`
`of the abdomen/pelvis, and radionuclide bone scan. To assess local disease, PCWG2
`
`suggests an endorectal MRI or ultrasound of the prostate or prostate bed. For those with
`
`symptoms of neurologic compromise, PCWG2 recommends MRI of the spine and base of
`
`the skull. PCWG2 also recognizes that detecting metastases will improve as more sensitive
`
`imaging tests become standard, but does not recommend positron emission tomography
`
`(PET) using fluorodeoxyglucose or other tracers and ProstaScint (Cytogen Corp, Princeton,
`
`NJ) scanning because they are considered investigational at this time.
`
`Symptoms and health-related quality of life—When enrolling a patient onto a
`
`clinical trial that incorporates symptoms and health-related quality of life, symptoms of
`
`disease should be characterized at baseline using validated instruments according to
`standards defined by the FDA in its guidance for patient-reported outcomes.8 The evaluation
`should include confirmation that patient input was included during development of the
`
`measure(s); that the content and the construct were validated; and that the measure(s) were
`
`reliable for the population being studied. A lead-in period of observation is advised to ensure
`
`adequate baseline assessments. Potentially relevant domains include pain, fatigue, anorexia/
`weight loss, constipation, and urinary symptoms.6,8
`
`The use of pain relief as a trial end point may be particularly valuable because the presence
`of pain is a known prognostic factor for survival,9 and palliation of symptoms is a
`therapeutic goal. An acceptable criterion for trial enrollment is new pain in an area of
`
`radiographically evident disease. Pain measures in particular should include assessments of
`
`intensity, frequency, and duration quantified (eg, on a five-point scale such as the McGill-
`
`Melzack Pain Questionnaire or IMMPACT [Initiative on Methods, Measurement, and Pain
`Assessment in Clinical Trials] recommendations).10 Level of bother, location(s), likely
`relationship to prostate cancer (v prior therapy or comorbidities), and analgesic requirements
`
`should also be recorded.
`
`Health-related quality of life can also be evaluated through patient self-reported instruments
`
`that have been developed in keeping with the FDA guidance. In addition to assessing
`
`selected symptoms, these instruments may emphasize the effects of disease on physical,
`
`social, psychological/emotional, and cognitive functioning. New instruments should also be
`
`developed, recognizing that validation is laborious and is generally outside the realm of a
`
`phase II trial.
`
`J Clin Oncol. Author manuscript; available in PMC 2014 May 05.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`MYLAN PHARMS. INC. EXHIBIT 1124 PAGE 6
`
`

`

`Scher et al.
`
`Page 7
`
`Criteria for disease progression—PCWG1 defined progression criteria for enrollment
`
`on the basis of changes in PSA, bone metastases, and measurable disease. PCWG2 retains
`
`most of the original recommendations with modifications (Table 2).
`
`PSA—For patients who manifested disease progression solely as a rising PSA level,
`
`PCWG1 required obtaining a sequence of rising values at least 1 week apart and made 5.0
`ng/mL the minimum starting level for trial entry (Table 2).1 PCWG2 keeps the timing of
`PSA testing at a minimum of 1-week intervals and recommends reducing the threshold PSA
`
`level from 5.0 ng/mL to 2.0 ng/mL because of the availability of more sensitive assays (Fig
`2). Given the prognostic significance of the rate of rise in PSA,11 PCWG2 advises
`estimating a pretreatment PSA-DT11a if at least three values are available, but does not
`recommend delaying either treatment or enrollment onto a trial simply to estimate PSA-
`DT.11a
`
`Target (nodal and visceral) lesions or measurable disease—A requirement of
`
`measurable lesions (target lesions as defined by RECIST) for trial entry is not recommended
`
`by PCWG2 because it shifts the emphasis from bone metastases, which develop in upwards
`
`of 90% of patients, to lymph nodes, which occur in only 20% to 25% of patients with
`prostate cancer and contribute less to morbidity than do other sites of metastases.3 The result
`is that much energy might be wasted on an end point of lesser clinical significance. That
`
`said, however, trials that are collecting data on measurable lesions should follow RECIST,
`
`and progression in a nodal or visceral site is sufficient to document disease progression.
`
`PCWG2 advises recording the presence or absence of nodal and visceral disease (ie, liver
`
`and lung) pretreatment and outcomes post-treatment separately. Up to 10 visceral and nodal
`lesions in total should be recorded (with a maximum of five in any one organ),2 although, in
`one series, the median number of target lesions was three.3 Because small lymph nodes are
`difficult to measure accurately and may not be malignant, PCWG2 recommends that the
`
`greatest diameter of a lymph node must measure at least 2 cm by spiral CT to be considered
`a target lesion.3
`
`The prostate (primary site)—PCWG2 recommends both recording the treatments that
`
`targeted the primary tumor and performing directed pelvic imaging to determine whether
`
`disease is present in this area at the time of enrollment. This should include a CT scan or
`
`MRI at a minimum, and, in centers with the relevant expertise, endorectal MRI or transrectal
`
`ultrasound. A prostate mass or recurrence in the prostate bed is not considered metastatic
`
`disease; many develop in cases where the surgical margins were positive or there was
`
`extracapsular extension.
`
`Bone—Even with the improved imaging modalities that have developed since the
`
`publication of PCWG1, it is difficult to interpret the clinical significance of changes in size
`
`or intensity of bone metastases on bone scan. When the bone scan is the sole indicator of
`
`progression, PCWG2 defines progression in bone when at least two or more new lesions are
`
`seen on bone scan compared with a prior scan for trial entry. In situations where the scan
`
`findings are suggestive of a flare reaction, or apparent new lesion(s) may represent trauma, it
`
`may prove useful to confirm these results with other imaging modalities such as MRI or
`
`J Clin Oncol. Author manuscript; available in PMC 2014 May 05.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`MYLAN PHARMS. INC. EXHIBIT 1124 PAGE 7
`
`

`

`Scher et al.
`
`Page 8
`
`fine-cut CT. Confirmation is generally not necessary if multiple new areas of uptake are
`
`observed. Consistent criteria for progression of disease in bone using PET and MRI are still
`
`under investigation.
`
`Other sites of disease—PCWG2 recommends that patients who meet other trial criteria
`
`may be enrolled if they have epidural disease that has been treated and there is no
`
`progression in the treated area. Aside from epidural lesions, other nontarget lesions
`
`considered by RECIST (including leptomeningeal spread, ascites, pleural/pericardial
`
`effusions, and abdominal masses not confirmed) are rare in prostate cancer.
`
`Evaluating hormonal status and prior systemic therapies—Equally important as
`
`pretreatment assessments and in defining disease progression is to evaluate and record
`
`historical factors that may affect sensitivity to treatment.
`
`Prior hormonal interventions—PCWG2 considers a “hormonal intervention” the
`
`addition or discontinuation of a hormonal therapy with therapeutic intent because of disease
`
`progression. Androgen depletion that is discontinued and restarted as part of a planned
`
`intermittent or cycling approach is considered a single intervention independent of the
`
`number of cycles. PCWG2 recommends that investigators record the prior hormonal
`
`interventions received by a patient by documenting the number, type, and duration of
`
`administration when available. Although most patients treated with gonadotropin-releasing
`
`hormone (GnRH) analog therapy initially or subsequently receive an antiandrogen, many do
`
`not. The duration of administration may be as short as 1 month in some and continuous in
`
`others. Because a range of outcomes have been reported for secondary hormonal
`
`manipulations. PCWG2 advises classifying tumors that are progressing with castrate levels
`of testosterone as “castration resistant”12; and not “hormone refractory” because many
`patients respond to second- and third-line hormonal therapies. Patients who are receiving
`
`antiandrogens as monotherapy and have noncastrate testosterone levels should receive
`
`testosterone-lowering therapy before being considered for trial.
`
`Serum testosterone levels—PCWG1 defined castrate status as a serum testosterone
`
`level of less than 50 ng/dL (< 1.7 nmol/L). It is now recognized that the measured level of
`testosterone in the blood may not accurately reflect intratumoral androgen levels,13,14,14a
`which are often sufficient to stimulate tumor growth. Sources include the adrenal glands or
`
`the tumor itself as a result of the upregulation of the genes involved in androgen
`synthesis.15,16 Recognizing that some patients have higher testosterone levels despite
`continued androgen depletion with GnRH analog therapy, and that total testosterone levels
`
`do not reflect bioavailable testosterone because of the variation in the levels of sex hormone-
`
`binding globulin and the laboratory-to-laboratory variation in the measurement of hormone
`
`levels, PCWG2 retains the maximal serum testosterone level of 50 ng/dL (1.7 nmol/L) for
`
`entry. PCWG2 also reaffirms that castrate status be maintained for any patient who has not
`
`undergone surgical orchiectomy by continued GnRH analog administration, recognizing
`
`that, in some patients, testosterone levels might remain suppressed if GnRH analog therapy
`
`were discontinued.
`
`J Clin Oncol. Author manuscript; available in PMC 2014 May 05.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`MYLAN PHARMS. INC. EXHIBIT 1124 PAGE 8
`
`

`

`Scher et al.
`
`Page 9
`
`Antiandrogen discontinuation responses—To avoid misattributing benefit to a study
`
`agent, PCWG1 required documenting progressive disease after discontinuing antiandrogen
`
`treatment. However, because the time to completely withdraw from the treatment can range
`
`from 4 to 8 weeks, depending on the half-life of the antiandrogen, many otherwise eligible
`
`patients were not included in clinical trials. Withdrawal responses typically occur in patients
`
`who are treated with combined androgen blockade (a GnRH analog or orchiectomy in
`
`combination with continuous antiandrogen) as initial therapy for a prolonged period of time,
`or who have responded to adding a peripheral antiandrogen as second-line therapy.17 For
`these situations, PCWG2 recommends evaluating patients for withdrawal responses.
`
`PCWG2 advises investigators not to wait to assess for a withdrawal response in patients who
`
`did not respond or who showed a decline in PSA for 3 months or less after an antiandrogen
`
`was administered as a second-line or later intervention.
`
`Prior nonhormonal therapies—All of the treatments for local disease (surgery,
`
`radiation therapy, and so on) should be recorded. Other systemic therapies (eg, first-line
`
`docetaxel and/or biologic agents) should also be described in detail, including the response,
`the reason for discontinuation, and the interval off treatment.18
`
`Clinical Subtypes Based on Patterns of Spread
`
`Investigators can enhance eligibility guidelines by defining clinical subtypes based on the
`
`patterns of spread. PCWG1 defined four patient cohorts: (1) progressive measurable disease;
`
`(2) progressive bone metastases; (3) increasing PSA and stable metastases; and (4)
`increasing PSA and no metastatic disease.1 This classification does not reflect the pattern of
`spread of disease, nor does it separate patients on the basis of prognosis, more favorable for
`
`patients in the rising-PSA–castrate state with no documented metastatic disease at present or
`
`in the past, and worse for patients with visceral disease. In response, PCWG2 defines five
`
`patient cohorts (Table 3) that include a poor prognostic group with visceral disease, a group
`
`with bone metastases with or without nodal metastases but no visceral organ disease, a
`
`group with nodal disease and no visceral or bone disease, an

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