1
|
Fang F, Tamura RN, Braun TM, Kidwell KM. Comparing dose levels to placebo using a continuous outcome in a small n, sequential, multiple assignment, randomized trial (snSMART). Stat Biopharm Res 2022. [DOI: 10.1080/19466315.2022.2118162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Affiliation(s)
- Fang Fang
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Roy N. Tamura
- Health Informatics Institute, University of South Florida, Tampa, Florida
| | - Thomas M. Braun
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Kelley M. Kidwell
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
2
|
Ko CC, Yeh LR, Kuo YT, Chen JH. Imaging biomarkers for evaluating tumor response: RECIST and beyond. Biomark Res 2021; 9:52. [PMID: 34215324 PMCID: PMC8252278 DOI: 10.1186/s40364-021-00306-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 06/10/2021] [Indexed: 12/12/2022] Open
Abstract
Response Evaluation Criteria in Solid Tumors (RECIST) is the gold standard for assessment of treatment response in solid tumors. Morphologic change of tumor size evaluated by RECIST is often correlated with survival length and has been considered as a surrogate endpoint of therapeutic efficacy. However, the detection of morphologic change alone may not be sufficient for assessing response to new anti-cancer medication in all solid tumors. During the past fifteen years, several molecular-targeted therapies and immunotherapies have emerged in cancer treatment which work by disrupting signaling pathways and inhibited cell growth. Tumor necrosis or lack of tumor progression is associated with a good therapeutic response even in the absence of tumor shrinkage. Therefore, the use of unmodified RECIST criteria to estimate morphological changes of tumor alone may not be sufficient to estimate tumor response for these new anti-cancer drugs. Several studies have reported the low reliability of RECIST in evaluating treatment response in different tumors such as hepatocellular carcinoma, lung cancer, prostate cancer, brain glioma, bone metastasis, and lymphoma. There is an increased need for new medical imaging biomarkers, considering the changes in tumor viability, metabolic activity, and attenuation, which are related to early tumor response. Promising imaging techniques, beyond RECIST, include dynamic contrast-enhanced computed tomography (CT) or magnetic resonance imaging (MRI), diffusion-weight imaging (DWI), magnetic resonance spectroscopy (MRS), and 18 F-fluorodeoxyglucose (FDG) positron emission tomography (PET). This review outlines the current RECIST with their limitations and the new emerging concepts of imaging biomarkers in oncology.
Collapse
Affiliation(s)
- Ching-Chung Ko
- Department of Medical Imaging, Chi Mei Medical Center, Tainan, Taiwan.,Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Lee-Ren Yeh
- Department of Radiology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Yu-Ting Kuo
- Department of Medical Imaging, Chi Mei Medical Center, Tainan, Taiwan.,Department of Medical Imaging, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Jeon-Hor Chen
- Department of Radiology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan. .,Tu & Yuan Center for Functional Onco-Imaging, Department of Radiological Sciences, University of California, 164 Irvine Hall, Irvine, CA, 92697 - 5020, USA.
| |
Collapse
|
3
|
Fang F, Hochstedler KA, Tamura RN, Braun TM, Kidwell KM. Bayesian methods to compare dose levels with placebo in a small n, sequential, multiple assignment, randomized trial. Stat Med 2020; 40:963-977. [PMID: 33216360 DOI: 10.1002/sim.8813] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/15/2020] [Accepted: 10/30/2020] [Indexed: 11/11/2022]
Abstract
Clinical trials studying treatments for rare diseases are challenging to design and conduct due to the limited number of patients eligible for the trial. One design used to address this challenge is the small n, sequential, multiple assignment, randomized trial (snSMART). We propose a new snSMART design that investigates the response rates of a drug tested at a low and high dose compared with placebo. Patients are randomized to an initial treatment (stage 1). In stage 2, patients are rerandomized, depending on their initial treatment and their response to that treatment in stage 1, to either the same or a different dose of treatment. Data from both stages are used to determine the efficacy of the active treatment. We present a Bayesian approach where information is borrowed between stage 1 and stage 2. We compare our approach to standard methods using only stage 1 data and a log-linear Poisson model that uses data from both stages where parameters are estimated using generalized estimating equations. We observe that the Bayesian method has smaller root-mean-square-error and 95% credible interval widths than standard methods in the tested scenarios. We conclude that it is advantageous to utilize data from both stages for a primary efficacy analysis and that the specific snSMART design shown here can be used in the registration of a drug for the treatment of rare diseases.
Collapse
Affiliation(s)
- Fang Fang
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Kimberly A Hochstedler
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Roy N Tamura
- Health Informatics Institute, University of South Florida, Tampa, Florida, USA
| | - Thomas M Braun
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Kelley M Kidwell
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
Grayling MJ, Dimairo M, Mander AP, Jaki TF. A Review of Perspectives on the Use of Randomization in Phase II Oncology Trials. J Natl Cancer Inst 2019; 111:1255-1262. [PMID: 31218346 PMCID: PMC6910171 DOI: 10.1093/jnci/djz126] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/05/2019] [Accepted: 06/12/2019] [Indexed: 12/21/2022] Open
Abstract
Historically, phase II oncology trials assessed a treatment's efficacy by examining its tumor response rate in a single-arm trial. Then, approximately 25 years ago, certain statistical and pharmacological considerations ignited a debate around whether randomized designs should be used instead. Here, based on an extensive literature review, we review the arguments on either side of this debate. In particular, we describe the numerous factors that relate to the reliance of single-arm trials on historical control data and detail the trial scenarios in which there was general agreement on preferential utilization of single-arm or randomized design frameworks, such as the use of single-arm designs when investigating treatments for rare cancers. We then summarize the latest figures on phase II oncology trial design, contrasting current design choices against historical recommendations on best practice. Ultimately, we find several ways in which the design of recently completed phase II trials does not appear to align with said recommendations. For example, despite advice to the contrary, only 66.2% of the assessed trials that employed progression-free survival as a primary or coprimary outcome used a randomized comparative design. In addition, we identify that just 28.2% of the considered randomized comparative trials came to a positive conclusion as opposed to 72.7% of the single-arm trials. We conclude by describing a selection of important issues influencing contemporary design, framing this discourse in light of current trends in phase II, such as the increased use of biomarkers and recent interest in novel adaptive designs.
Collapse
Affiliation(s)
- Michael J Grayling
- Correspondence to: Michael J. Grayling, Institute of Health & Society, Newcastle University, Baddiley-Clark Building, Richardson Rd, Newcastle upon Tyne NE2 4AX, UK (e-mail: )
| | | | | | | |
Collapse
|
5
|
Jones RL, Ratain MJ, O'Dwyer PJ, Siu LL, Jassem J, Medioni J, DeJonge M, Rudin C, Sawyer M, Khayat D, Awada A, de Vos-Geelen JMPGM, Evans TRJ, Obel J, Brockstein B, DeGreve J, Baurain JF, Maki R, D'Adamo D, Dickson M, Undevia S, Geary D, Janisch L, Bedard PL, Abdul Razak AR, Kristeleit R, Vitfell-Rasmussen J, Walters I, Kaye SB, Schwartz G. Phase II randomised discontinuation trial of brivanib in patients with advanced solid tumours. Eur J Cancer 2019; 120:132-139. [PMID: 31522033 PMCID: PMC8852771 DOI: 10.1016/j.ejca.2019.07.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/20/2019] [Accepted: 07/23/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND Brivanib is a selective inhibitor of vascular endothelial growth factor and fibroblast growth factor (FGF) signalling. We performed a phase II randomised discontinuation trial of brivanib in 7 tumour types (soft-tissue sarcomas [STS], ovarian cancer, breast cancer, pancreatic cancer, non-small-cell lung cancer [NSCLC], gastric/esophageal cancer and transitional cell carcinoma [TCC]). PATIENTS AND METHODS During a 12-week open-label lead-in period, patients received brivanib 800 mg daily and were evaluated for FGF2 status by immunohistochemistry. Patients with stable disease at week 12 were randomised to brivanib or placebo. A study steering committee evaluated week 12 response to determine if enrolment in a tumour type would continue. The primary objective was progression-free survival (PFS) for brivanib versus placebo in patients with FGF2-positive tumours. RESULTS A total of 595 patients were treated, and stable disease was observed at the week 12 randomisation point in all tumour types. Closure decisions were made for breast cancer, pancreatic cancer, NSCLC, gastric cancer and TCC. Criteria for expansion were met for STS and ovarian cancer. In 53 randomised patients with STS and FGF2-positive tumours, the median PFS was 2.8 months for brivanib and 1.4 months for placebo (hazard ratio [HR]: 0.58, p = 0.08). For all randomised patients with sarcomas, the median PFS was 2.8 months (95% confidence interval [CI]: 1.4-4.0) for those treated with brivanib compared with 1.4 months (95% CI: 1.3-1.6) for placebo (HR = 0.64, 95% CI: 0.38-1.07; p = 0.09). In the 36 randomised patients with ovarian cancer and FGF2-positive tumours, the median PFS was 4.0 (95% CI: 2.6-4.2) months for brivanib and 2.0 months (95% CI: 1.2-2.7) for placebo (HR: 0.56, 95% CI: 0.26-1.22). For all randomised patients with ovarian cancer, the median PFS in those randomised to brivanib was 4.0 months (95% CI: 2.6-4.2) and was 2.0 months (95% CI: 1.2-2.7) in those randomised to placebo (HR = 0.54, 95% CI: 0.25-1.17; p = 0.11). CONCLUSION Brivanib demonstrated activity in STS and ovarian cancer with an acceptable safety profile. FGF2 expression, as defined in the protocol, is not a predictive biomarker of the efficacy of brivanib.
Collapse
Affiliation(s)
- Robin L Jones
- Royal Marsden Hospital, Institute of Cancer Research, London, United Kingdom.
| | | | | | | | | | - Jacques Medioni
- Hôpital Européen Georges Pompidou, Paris, France; Paris-Descartes University, Paris, France
| | - Maja DeJonge
- Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | | | | | | | - Judith M P G M de Vos-Geelen
- Department of Internal Medicine, Division of Medical Oncology, GROW - School for Oncology and Developmental Biology, Maastricht UMC+, Maastricht, the Netherlands
| | - T R Jeffry Evans
- Beatson West of Scotland Cancer Centre, University of Glasgow, Glasgow, United Kingdom
| | - Jennifer Obel
- North Shore University Health System, Evanston, IL, USA
| | | | | | | | | | - David D'Adamo
- Eisai Inc, Woodcliff Lake, NJ Previously Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Mark Dickson
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | | | - Rebecca Kristeleit
- Royal Marsden Hospital, Institute of Cancer Research, London, United Kingdom
| | | | - Ian Walters
- Intensity Therapeutics Inc, Westport, CT Previously BMS, USA
| | - Stan B Kaye
- Royal Marsden Hospital, Institute of Cancer Research, London, United Kingdom
| | | |
Collapse
|
6
|
Liu M, Dressler EV. A predictive probability interim design for phase II clinical trials with continuous endpoints. Stat Med 2018; 37:1960-1972. [PMID: 29611211 DOI: 10.1002/sim.7659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 02/11/2018] [Accepted: 02/15/2018] [Indexed: 11/07/2022]
Abstract
Molecular targeted therapies come often with lower toxicity profiles than traditional cytotoxic treatments, thus shifting drug development paradigm into establishing evidence of biological activity, target modulation, and pharmacodynamics effects of these therapies in early phase trials. Therefore, these trials need to address simultaneous evaluation of safety, proof-of-concept biological marker activity, or changes in continuous tumor size instead of binary response rate. Interim analyses are typically incorporated in the trial due to concerns regarding excessive toxicity and ineffective new treatment. There is a lack of interim strategies developed to monitor futility and/or efficacy for these types of continuous outcomes, especially in single-arm phase II trials. We propose a 2-stage design based on predictive probability to accommodate continuous endpoints, assuming a normal distribution with known variance. Simulation results and case study demonstrated that the proposed design can incorporate an interim stop for futility as well as for efficacy while maintaining desirable design properties. As expected, using continuous tumor size resulted in reduced sample sizes for both optimal and minimax designs. A limited exploration of various priors was performed and shown to be robust. As research rapidly moves to incorporate more molecular targeted therapies, it will accommodate new types of outcomes while allowing for flexible stopping rules to continue optimizing trial resources and prioritize agents with compelling early phase data.
Collapse
Affiliation(s)
- Meng Liu
- Department of Biostatistics, University of Kentucky, Lexington, KY, U.S.A
| | - Emily V Dressler
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, U.S.A
| |
Collapse
|
7
|
Response evaluation of giant-cell tumor of bone treated by denosumab: Histogram and texture analysis of CT images. J Orthop Sci 2018; 23:570-577. [PMID: 29429890 DOI: 10.1016/j.jos.2018.01.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 01/15/2018] [Accepted: 01/16/2018] [Indexed: 01/07/2023]
Abstract
BACKGROUND This study aimed to compare computed tomography (CT) features, including tumor size and textural and histogram measurements, of giant-cell tumors of bone (GCTBs) before and after denosumab treatment and determine their applicability in monitoring GCTB response to denosumab treatment. METHODS This retrospective study included eight patients (male, 3; female, 5; mean age, 33.4 years) diagnosed with GCTB, who had received treatment by denosumab and had undergone pre- and post-treatment non-contrast CT between January 2010 and December 2016. This study was approved by the institutional review board. Pre- and post-treatment size, histogram, and textural parameters of GCTBs were compared by the Wilcoxon signed-rank test. Pathological findings of five patients who underwent surgery after denosumab treatment were evaluated for assessment of treatment response. RESULTS Relative to the baseline values, the tumor size had decreased, while the mean attenuation, standard deviation, entropy (all, P = 0.017), and skewness (P = 0.036) of the GCTBs had significantly increased post-treatment. Although the difference was statistically insignificant, the tumors also exhibited increased kurtosis, contrast, and inverse difference moment (P = 0.123, 0.327, and 0.575, respectively) post-treatment. Histologic findings revealed new bone formation and complete depletion or decrease in the number of osteoclast-like giant cells. CONCLUSION The histogram and textural parameters of GCTBs changed significantly after denosumab treatment. Knowledge of the tendency towards increased mean attenuation and heterogeneity but increased local homogeneity in post-treatment CT histogram and textural features of GCTBs might aid in treatment planning and tumor response evaluation during denosumab treatment.
Collapse
|
8
|
Schadendorf D, Wolchok JD, Hodi FS, Chiarion-Sileni V, Gonzalez R, Rutkowski P, Grob JJ, Cowey CL, Lao CD, Chesney J, Robert C, Grossmann K, McDermott D, Walker D, Bhore R, Larkin J, Postow MA. Efficacy and Safety Outcomes in Patients With Advanced Melanoma Who Discontinued Treatment With Nivolumab and Ipilimumab Because of Adverse Events: A Pooled Analysis of Randomized Phase II and III Trials. J Clin Oncol 2017; 35:3807-3814. [PMID: 28841387 PMCID: PMC5791828 DOI: 10.1200/jco.2017.73.2289] [Citation(s) in RCA: 325] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Purpose Approximately 40% of patients with advanced melanoma who received nivolumab combined with ipilimumab in clinical trials discontinued treatment because of adverse events (AEs). We conducted a retrospective analysis to assess the efficacy and safety of nivolumab plus ipilimumab in patients who discontinued treatment because of AEs. Methods Data were pooled from phase II and III trials of patients who received nivolumab 1 mg/kg plus ipilimumab 3 mg/kg, every 3 weeks for four doses, followed by nivolumab monotherapy 3 mg/kg every 2 weeks (N = 409). Efficacy was assessed in all randomly assigned patients who discontinued because of AEs during the induction phase (n = 96) and in those who did not discontinue because of AEs (n = 233). Safety was assessed in treated patients who discontinued because of AEs (n = 176) at any time and in those who did not discontinue because of AEs (n = 231). Results At a minimum follow-up of 18 months, median progression-free survival was 8.4 months for patients who discontinued treatment because of AEs during the induction phase and 10.8 months for patients who did not discontinue because of AEs ( P = .97). Median overall survival had not been reached in either group ( P = .23). The objective response rate was 58.3% for patients who discontinued because of AEs during the induction phase and 50.2% for patients who did not discontinue. The vast majority of grade 3 or 4 AEs occurred during the induction phase, with most resolving after appropriate management. Conclusion Efficacy outcomes seemed similar between patients who discontinued nivolumab plus ipilimumab treatment because of AEs during the induction phase and those who did not discontinue because of AEs. Therefore, even after discontinuation, many patients may continue to derive benefit from combination therapy.
Collapse
Affiliation(s)
- Dirk Schadendorf
- Dirk Schadendorf, University Hospital Essen and the German Cancer Consortium, Essen, Germany; Jedd D. Wolchok and Michael A. Postow, Memorial Sloan Kettering Cancer Center; Michael A. Postow, Weill Cornell Medical College, New York, NY; F. Stephen Hodi, Dana-Farber Cancer Institute; David McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Vanna Chiarion-Sileni, Istituto Oncologico Veneto, Veneto, Italy; Rene Gonzalez, University of Colorado Denver, Aurora, CO; Piotr Rutkowski, Maria Skłodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jean-Jacques Grob, Aix-Marseille Université, Assistance Publique Hôpitaux de Marseille Timone, Marseille; Caroline Robert, Gustave Roussy and Université Paris-Sud, Paris, France; C. Lance Cowey, Texas Oncology-Baylor Cancer Center, Dallas, TX; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Jason Chesney, University of Louisville, Louisville, KY; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Dana Walker and Rafia Bhore, Bristol-Myers Squibb, Princeton, NJ; and James Larkin, Royal Marsden Hospital, London, United Kingdom
| | - Jedd D. Wolchok
- Dirk Schadendorf, University Hospital Essen and the German Cancer Consortium, Essen, Germany; Jedd D. Wolchok and Michael A. Postow, Memorial Sloan Kettering Cancer Center; Michael A. Postow, Weill Cornell Medical College, New York, NY; F. Stephen Hodi, Dana-Farber Cancer Institute; David McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Vanna Chiarion-Sileni, Istituto Oncologico Veneto, Veneto, Italy; Rene Gonzalez, University of Colorado Denver, Aurora, CO; Piotr Rutkowski, Maria Skłodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jean-Jacques Grob, Aix-Marseille Université, Assistance Publique Hôpitaux de Marseille Timone, Marseille; Caroline Robert, Gustave Roussy and Université Paris-Sud, Paris, France; C. Lance Cowey, Texas Oncology-Baylor Cancer Center, Dallas, TX; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Jason Chesney, University of Louisville, Louisville, KY; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Dana Walker and Rafia Bhore, Bristol-Myers Squibb, Princeton, NJ; and James Larkin, Royal Marsden Hospital, London, United Kingdom
| | - F. Stephen Hodi
- Dirk Schadendorf, University Hospital Essen and the German Cancer Consortium, Essen, Germany; Jedd D. Wolchok and Michael A. Postow, Memorial Sloan Kettering Cancer Center; Michael A. Postow, Weill Cornell Medical College, New York, NY; F. Stephen Hodi, Dana-Farber Cancer Institute; David McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Vanna Chiarion-Sileni, Istituto Oncologico Veneto, Veneto, Italy; Rene Gonzalez, University of Colorado Denver, Aurora, CO; Piotr Rutkowski, Maria Skłodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jean-Jacques Grob, Aix-Marseille Université, Assistance Publique Hôpitaux de Marseille Timone, Marseille; Caroline Robert, Gustave Roussy and Université Paris-Sud, Paris, France; C. Lance Cowey, Texas Oncology-Baylor Cancer Center, Dallas, TX; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Jason Chesney, University of Louisville, Louisville, KY; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Dana Walker and Rafia Bhore, Bristol-Myers Squibb, Princeton, NJ; and James Larkin, Royal Marsden Hospital, London, United Kingdom
| | - Vanna Chiarion-Sileni
- Dirk Schadendorf, University Hospital Essen and the German Cancer Consortium, Essen, Germany; Jedd D. Wolchok and Michael A. Postow, Memorial Sloan Kettering Cancer Center; Michael A. Postow, Weill Cornell Medical College, New York, NY; F. Stephen Hodi, Dana-Farber Cancer Institute; David McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Vanna Chiarion-Sileni, Istituto Oncologico Veneto, Veneto, Italy; Rene Gonzalez, University of Colorado Denver, Aurora, CO; Piotr Rutkowski, Maria Skłodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jean-Jacques Grob, Aix-Marseille Université, Assistance Publique Hôpitaux de Marseille Timone, Marseille; Caroline Robert, Gustave Roussy and Université Paris-Sud, Paris, France; C. Lance Cowey, Texas Oncology-Baylor Cancer Center, Dallas, TX; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Jason Chesney, University of Louisville, Louisville, KY; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Dana Walker and Rafia Bhore, Bristol-Myers Squibb, Princeton, NJ; and James Larkin, Royal Marsden Hospital, London, United Kingdom
| | - Rene Gonzalez
- Dirk Schadendorf, University Hospital Essen and the German Cancer Consortium, Essen, Germany; Jedd D. Wolchok and Michael A. Postow, Memorial Sloan Kettering Cancer Center; Michael A. Postow, Weill Cornell Medical College, New York, NY; F. Stephen Hodi, Dana-Farber Cancer Institute; David McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Vanna Chiarion-Sileni, Istituto Oncologico Veneto, Veneto, Italy; Rene Gonzalez, University of Colorado Denver, Aurora, CO; Piotr Rutkowski, Maria Skłodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jean-Jacques Grob, Aix-Marseille Université, Assistance Publique Hôpitaux de Marseille Timone, Marseille; Caroline Robert, Gustave Roussy and Université Paris-Sud, Paris, France; C. Lance Cowey, Texas Oncology-Baylor Cancer Center, Dallas, TX; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Jason Chesney, University of Louisville, Louisville, KY; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Dana Walker and Rafia Bhore, Bristol-Myers Squibb, Princeton, NJ; and James Larkin, Royal Marsden Hospital, London, United Kingdom
| | - Piotr Rutkowski
- Dirk Schadendorf, University Hospital Essen and the German Cancer Consortium, Essen, Germany; Jedd D. Wolchok and Michael A. Postow, Memorial Sloan Kettering Cancer Center; Michael A. Postow, Weill Cornell Medical College, New York, NY; F. Stephen Hodi, Dana-Farber Cancer Institute; David McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Vanna Chiarion-Sileni, Istituto Oncologico Veneto, Veneto, Italy; Rene Gonzalez, University of Colorado Denver, Aurora, CO; Piotr Rutkowski, Maria Skłodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jean-Jacques Grob, Aix-Marseille Université, Assistance Publique Hôpitaux de Marseille Timone, Marseille; Caroline Robert, Gustave Roussy and Université Paris-Sud, Paris, France; C. Lance Cowey, Texas Oncology-Baylor Cancer Center, Dallas, TX; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Jason Chesney, University of Louisville, Louisville, KY; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Dana Walker and Rafia Bhore, Bristol-Myers Squibb, Princeton, NJ; and James Larkin, Royal Marsden Hospital, London, United Kingdom
| | - Jean-Jacques Grob
- Dirk Schadendorf, University Hospital Essen and the German Cancer Consortium, Essen, Germany; Jedd D. Wolchok and Michael A. Postow, Memorial Sloan Kettering Cancer Center; Michael A. Postow, Weill Cornell Medical College, New York, NY; F. Stephen Hodi, Dana-Farber Cancer Institute; David McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Vanna Chiarion-Sileni, Istituto Oncologico Veneto, Veneto, Italy; Rene Gonzalez, University of Colorado Denver, Aurora, CO; Piotr Rutkowski, Maria Skłodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jean-Jacques Grob, Aix-Marseille Université, Assistance Publique Hôpitaux de Marseille Timone, Marseille; Caroline Robert, Gustave Roussy and Université Paris-Sud, Paris, France; C. Lance Cowey, Texas Oncology-Baylor Cancer Center, Dallas, TX; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Jason Chesney, University of Louisville, Louisville, KY; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Dana Walker and Rafia Bhore, Bristol-Myers Squibb, Princeton, NJ; and James Larkin, Royal Marsden Hospital, London, United Kingdom
| | - C. Lance Cowey
- Dirk Schadendorf, University Hospital Essen and the German Cancer Consortium, Essen, Germany; Jedd D. Wolchok and Michael A. Postow, Memorial Sloan Kettering Cancer Center; Michael A. Postow, Weill Cornell Medical College, New York, NY; F. Stephen Hodi, Dana-Farber Cancer Institute; David McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Vanna Chiarion-Sileni, Istituto Oncologico Veneto, Veneto, Italy; Rene Gonzalez, University of Colorado Denver, Aurora, CO; Piotr Rutkowski, Maria Skłodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jean-Jacques Grob, Aix-Marseille Université, Assistance Publique Hôpitaux de Marseille Timone, Marseille; Caroline Robert, Gustave Roussy and Université Paris-Sud, Paris, France; C. Lance Cowey, Texas Oncology-Baylor Cancer Center, Dallas, TX; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Jason Chesney, University of Louisville, Louisville, KY; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Dana Walker and Rafia Bhore, Bristol-Myers Squibb, Princeton, NJ; and James Larkin, Royal Marsden Hospital, London, United Kingdom
| | - Christopher D. Lao
- Dirk Schadendorf, University Hospital Essen and the German Cancer Consortium, Essen, Germany; Jedd D. Wolchok and Michael A. Postow, Memorial Sloan Kettering Cancer Center; Michael A. Postow, Weill Cornell Medical College, New York, NY; F. Stephen Hodi, Dana-Farber Cancer Institute; David McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Vanna Chiarion-Sileni, Istituto Oncologico Veneto, Veneto, Italy; Rene Gonzalez, University of Colorado Denver, Aurora, CO; Piotr Rutkowski, Maria Skłodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jean-Jacques Grob, Aix-Marseille Université, Assistance Publique Hôpitaux de Marseille Timone, Marseille; Caroline Robert, Gustave Roussy and Université Paris-Sud, Paris, France; C. Lance Cowey, Texas Oncology-Baylor Cancer Center, Dallas, TX; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Jason Chesney, University of Louisville, Louisville, KY; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Dana Walker and Rafia Bhore, Bristol-Myers Squibb, Princeton, NJ; and James Larkin, Royal Marsden Hospital, London, United Kingdom
| | - Jason Chesney
- Dirk Schadendorf, University Hospital Essen and the German Cancer Consortium, Essen, Germany; Jedd D. Wolchok and Michael A. Postow, Memorial Sloan Kettering Cancer Center; Michael A. Postow, Weill Cornell Medical College, New York, NY; F. Stephen Hodi, Dana-Farber Cancer Institute; David McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Vanna Chiarion-Sileni, Istituto Oncologico Veneto, Veneto, Italy; Rene Gonzalez, University of Colorado Denver, Aurora, CO; Piotr Rutkowski, Maria Skłodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jean-Jacques Grob, Aix-Marseille Université, Assistance Publique Hôpitaux de Marseille Timone, Marseille; Caroline Robert, Gustave Roussy and Université Paris-Sud, Paris, France; C. Lance Cowey, Texas Oncology-Baylor Cancer Center, Dallas, TX; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Jason Chesney, University of Louisville, Louisville, KY; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Dana Walker and Rafia Bhore, Bristol-Myers Squibb, Princeton, NJ; and James Larkin, Royal Marsden Hospital, London, United Kingdom
| | - Caroline Robert
- Dirk Schadendorf, University Hospital Essen and the German Cancer Consortium, Essen, Germany; Jedd D. Wolchok and Michael A. Postow, Memorial Sloan Kettering Cancer Center; Michael A. Postow, Weill Cornell Medical College, New York, NY; F. Stephen Hodi, Dana-Farber Cancer Institute; David McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Vanna Chiarion-Sileni, Istituto Oncologico Veneto, Veneto, Italy; Rene Gonzalez, University of Colorado Denver, Aurora, CO; Piotr Rutkowski, Maria Skłodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jean-Jacques Grob, Aix-Marseille Université, Assistance Publique Hôpitaux de Marseille Timone, Marseille; Caroline Robert, Gustave Roussy and Université Paris-Sud, Paris, France; C. Lance Cowey, Texas Oncology-Baylor Cancer Center, Dallas, TX; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Jason Chesney, University of Louisville, Louisville, KY; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Dana Walker and Rafia Bhore, Bristol-Myers Squibb, Princeton, NJ; and James Larkin, Royal Marsden Hospital, London, United Kingdom
| | - Kenneth Grossmann
- Dirk Schadendorf, University Hospital Essen and the German Cancer Consortium, Essen, Germany; Jedd D. Wolchok and Michael A. Postow, Memorial Sloan Kettering Cancer Center; Michael A. Postow, Weill Cornell Medical College, New York, NY; F. Stephen Hodi, Dana-Farber Cancer Institute; David McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Vanna Chiarion-Sileni, Istituto Oncologico Veneto, Veneto, Italy; Rene Gonzalez, University of Colorado Denver, Aurora, CO; Piotr Rutkowski, Maria Skłodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jean-Jacques Grob, Aix-Marseille Université, Assistance Publique Hôpitaux de Marseille Timone, Marseille; Caroline Robert, Gustave Roussy and Université Paris-Sud, Paris, France; C. Lance Cowey, Texas Oncology-Baylor Cancer Center, Dallas, TX; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Jason Chesney, University of Louisville, Louisville, KY; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Dana Walker and Rafia Bhore, Bristol-Myers Squibb, Princeton, NJ; and James Larkin, Royal Marsden Hospital, London, United Kingdom
| | - David McDermott
- Dirk Schadendorf, University Hospital Essen and the German Cancer Consortium, Essen, Germany; Jedd D. Wolchok and Michael A. Postow, Memorial Sloan Kettering Cancer Center; Michael A. Postow, Weill Cornell Medical College, New York, NY; F. Stephen Hodi, Dana-Farber Cancer Institute; David McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Vanna Chiarion-Sileni, Istituto Oncologico Veneto, Veneto, Italy; Rene Gonzalez, University of Colorado Denver, Aurora, CO; Piotr Rutkowski, Maria Skłodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jean-Jacques Grob, Aix-Marseille Université, Assistance Publique Hôpitaux de Marseille Timone, Marseille; Caroline Robert, Gustave Roussy and Université Paris-Sud, Paris, France; C. Lance Cowey, Texas Oncology-Baylor Cancer Center, Dallas, TX; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Jason Chesney, University of Louisville, Louisville, KY; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Dana Walker and Rafia Bhore, Bristol-Myers Squibb, Princeton, NJ; and James Larkin, Royal Marsden Hospital, London, United Kingdom
| | - Dana Walker
- Dirk Schadendorf, University Hospital Essen and the German Cancer Consortium, Essen, Germany; Jedd D. Wolchok and Michael A. Postow, Memorial Sloan Kettering Cancer Center; Michael A. Postow, Weill Cornell Medical College, New York, NY; F. Stephen Hodi, Dana-Farber Cancer Institute; David McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Vanna Chiarion-Sileni, Istituto Oncologico Veneto, Veneto, Italy; Rene Gonzalez, University of Colorado Denver, Aurora, CO; Piotr Rutkowski, Maria Skłodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jean-Jacques Grob, Aix-Marseille Université, Assistance Publique Hôpitaux de Marseille Timone, Marseille; Caroline Robert, Gustave Roussy and Université Paris-Sud, Paris, France; C. Lance Cowey, Texas Oncology-Baylor Cancer Center, Dallas, TX; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Jason Chesney, University of Louisville, Louisville, KY; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Dana Walker and Rafia Bhore, Bristol-Myers Squibb, Princeton, NJ; and James Larkin, Royal Marsden Hospital, London, United Kingdom
| | - Rafia Bhore
- Dirk Schadendorf, University Hospital Essen and the German Cancer Consortium, Essen, Germany; Jedd D. Wolchok and Michael A. Postow, Memorial Sloan Kettering Cancer Center; Michael A. Postow, Weill Cornell Medical College, New York, NY; F. Stephen Hodi, Dana-Farber Cancer Institute; David McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Vanna Chiarion-Sileni, Istituto Oncologico Veneto, Veneto, Italy; Rene Gonzalez, University of Colorado Denver, Aurora, CO; Piotr Rutkowski, Maria Skłodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jean-Jacques Grob, Aix-Marseille Université, Assistance Publique Hôpitaux de Marseille Timone, Marseille; Caroline Robert, Gustave Roussy and Université Paris-Sud, Paris, France; C. Lance Cowey, Texas Oncology-Baylor Cancer Center, Dallas, TX; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Jason Chesney, University of Louisville, Louisville, KY; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Dana Walker and Rafia Bhore, Bristol-Myers Squibb, Princeton, NJ; and James Larkin, Royal Marsden Hospital, London, United Kingdom
| | - James Larkin
- Dirk Schadendorf, University Hospital Essen and the German Cancer Consortium, Essen, Germany; Jedd D. Wolchok and Michael A. Postow, Memorial Sloan Kettering Cancer Center; Michael A. Postow, Weill Cornell Medical College, New York, NY; F. Stephen Hodi, Dana-Farber Cancer Institute; David McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Vanna Chiarion-Sileni, Istituto Oncologico Veneto, Veneto, Italy; Rene Gonzalez, University of Colorado Denver, Aurora, CO; Piotr Rutkowski, Maria Skłodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jean-Jacques Grob, Aix-Marseille Université, Assistance Publique Hôpitaux de Marseille Timone, Marseille; Caroline Robert, Gustave Roussy and Université Paris-Sud, Paris, France; C. Lance Cowey, Texas Oncology-Baylor Cancer Center, Dallas, TX; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Jason Chesney, University of Louisville, Louisville, KY; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Dana Walker and Rafia Bhore, Bristol-Myers Squibb, Princeton, NJ; and James Larkin, Royal Marsden Hospital, London, United Kingdom
| | - Michael A. Postow
- Dirk Schadendorf, University Hospital Essen and the German Cancer Consortium, Essen, Germany; Jedd D. Wolchok and Michael A. Postow, Memorial Sloan Kettering Cancer Center; Michael A. Postow, Weill Cornell Medical College, New York, NY; F. Stephen Hodi, Dana-Farber Cancer Institute; David McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Vanna Chiarion-Sileni, Istituto Oncologico Veneto, Veneto, Italy; Rene Gonzalez, University of Colorado Denver, Aurora, CO; Piotr Rutkowski, Maria Skłodowska Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Jean-Jacques Grob, Aix-Marseille Université, Assistance Publique Hôpitaux de Marseille Timone, Marseille; Caroline Robert, Gustave Roussy and Université Paris-Sud, Paris, France; C. Lance Cowey, Texas Oncology-Baylor Cancer Center, Dallas, TX; Christopher D. Lao, University of Michigan, Ann Arbor, MI; Jason Chesney, University of Louisville, Louisville, KY; Kenneth Grossmann, Huntsman Cancer Institute, Salt Lake City, UT; Dana Walker and Rafia Bhore, Bristol-Myers Squibb, Princeton, NJ; and James Larkin, Royal Marsden Hospital, London, United Kingdom
| |
Collapse
|
9
|
Ray-Coquard I, Trama A, Seckl MJ, Fotopoulou C, Pautier P, Pignata S, Kristensen G, Mangili G, Falconer H, Massuger L, Sehouli J, Pujade-Lauraine E, Lorusso D, Amant F, Rokkones E, Vergote I, Ledermann JA. Rare ovarian tumours: Epidemiology, treatment challenges in and outside a network setting. Eur J Surg Oncol 2017; 45:67-74. [PMID: 29108961 DOI: 10.1016/j.ejso.2017.09.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/26/2017] [Accepted: 09/30/2017] [Indexed: 10/18/2022] Open
Abstract
PURPOSE OF THE REVIEW More than 50% of all gynaecological cancers can be classified as rare tumours (defined as an annual incidence of <6 per 100,000) and such tumours represent an important challenge for clinicians. RECENT FINDINGS Rare cancers account for more than one fifth of all new cancer diagnoses, more than any of the single common cancers alone. Reviewing the RARECAREnet database, some of the tumours occur infrequently, whilst others because of their natural history have a high prevalence, and therefore appear to be more common, although their incidence is also rare. Harmonization of medical practice, guidelines and novel trials are needed to identify rare tumours and facilitate the development of new treatments. Ovarian tumours are the focus of this review, but we comment on other rare gynaecological tumours, as the diagnosis and treatment challenges faced are similar. FUTURE This requires European collaboration, international partnerships, harmonization of treatment and collaboration to overcome the regulatory barriers to conduct international trials. Whilst randomized trials can be done in many tumour types, there are some for which conducting even single arm studies may be challenging. For these tumours alternative study designs, robust collection of data through national registries and audits could lead to improvements in the treatment of rare tumours. In addition, concentring the care of patients with rare tumours into a limited number of centres will help to build expertise, facilitate trials and improve outcomes.
Collapse
Affiliation(s)
- I Ray-Coquard
- Dpt of Medical Oncology, Centre Leon Berard, University Claude Bernard LyonI, Lyon, France.
| | - AnnaLisa Trama
- AnnaLisa Trama, Fondazione IRCCS istituto nazionale dei tumori Milan, Italy
| | - M J Seckl
- Charing Cross Hospital, Campus of Imperial College London, Fulham Palace Rd, W68RF London, UK
| | - C Fotopoulou
- Dept of Surgery and Cancer, Imperial College London, UK
| | - P Pautier
- Medical Oncology, Dpt Gustave Roussy Institution, Villejuif, France
| | - S Pignata
- Medical Oncology, Department of Urology and Gynecology, Istituto Nazionale Tumori - IRCSS - Fondazione G. Pascale, Naples Italy
| | - G Kristensen
- Dept of Gynecologic Oncology, Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - G Mangili
- Department of Obstetrics and Gynecology, San Raffaele Scientific Institute, Milan, Italy
| | - H Falconer
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet/University Hospital, 171 76 Stockholm, Sweden
| | - L Massuger
- Department of Obstetrics and Gynaecology, Radboudumc, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - J Sehouli
- Department of Gynecology with Center for Oncological Surgery, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Medical University of Berlin, Germany
| | | | - D Lorusso
- Gynecologic Oncology Unit, Fondazione IRCCS istituto nazionale dei tumori Milan, Italy
| | - F Amant
- Center Gynaecologic Oncology Amsterdam (CGOA), Netherlands Cancer Institute, University of Amsterdam & Gynaecologic Oncology KU Leuven, The Netherlands
| | - E Rokkones
- Dept. of Gynaecological Oncology, The Norwegian Radium Hospital, Division of Cancer Medicine Oslo University Hospital, PO Box 4950 Nydalen, 0424 Oslo, Norway
| | - I Vergote
- Gynaecological Oncologist, University Hospital Leuven, European Union, Herestraat 49, B-3000 Leuven, Belgium
| | | | | |
Collapse
|
10
|
Schöffski P, Gordon M, Smith DC, Kurzrock R, Daud A, Vogelzang NJ, Lee Y, Scheffold C, Shapiro GI. Phase II randomised discontinuation trial of cabozantinib in patients with advanced solid tumours. Eur J Cancer 2017; 86:296-304. [PMID: 29059635 DOI: 10.1016/j.ejca.2017.09.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 09/14/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cabozantinib is an inhibitor of tyrosine kinases, including MET, vascular endothelial growth factor receptor, AXL and RET. This multi-cohort phase II randomised discontinuation trial explored anticancer activity of cabozantinib in nine tumour types. PATIENTS AND METHODS Cabozantinib was administered (100 mg, once daily) to patients with advanced, recurrent or metastatic cancers. Those with stable disease at week 12 were randomised 1:1 to cabozantinib or placebo. Primary end-points were objective response rate (ORR) at week 12 and progression-free survival (PFS) in the randomised phase. RESULTS A total of 526 patients were enrolled. The highest ORR was observed in ovarian cancer (OC) (21.7%); the largest PFS benefit was observed in castration-resistant prostate cancer (CRPC) (median 5.5 versus 1.4 months for placebo; hazard ratio 0.14, 95% confidence interval: 0.04, 0.52). Disease control rates were >40% for CRPC, OC, melanoma, metastatic breast cancer (MBC), hepatocellular carcinoma (HCC) and non-small cell lung cancer. Median duration of response ranged from 3.3 (MBC) to 11.2 months (OC). Encouraging efficacy results and symptomatic improvements prompted early suspension of the randomised stage and conversion to open-label non-randomised expansion cohorts. Dose reductions to manage adverse events (AEs) occurred in 48.7% of patients. The most frequent grade III-IV AEs were fatigue (12.4%), diarrhoea (10.5%), hypertension (10.5%) and palmar-plantar erythrodysesthesia syndrome (8.7%). CONCLUSIONS Clinical antitumour activity of cabozantinib was observed in a subset of tumour types: CRPC and OC were evaluated further in expansion cohorts. Phase III programs were initiated in CRPC and HCC. Interpretation of efficacy outcomes was limited by early termination of the randomised portion of the trial. TRIAL REGISTRATION NUMBER NCT00940225.
Collapse
Affiliation(s)
- Patrick Schöffski
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospital Leuven, Leuven, Belgium.
| | | | - David C Smith
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA.
| | - Razelle Kurzrock
- Moores Cancer Center, University of California, San Diego, San Diego, CA, USA.
| | - Adil Daud
- Medical Center at Parnassus, University of California, San Francisco, San Francisco, CA, USA.
| | - Nicholas J Vogelzang
- Comprehensive Cancer Centers of Nevada, US Oncology Research, Las Vegas, NV, USA.
| | - Yihua Lee
- Exelixis, Inc., South San Francisco, CA, USA.
| | | | - Geoffrey I Shapiro
- Early Drug Development Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
11
|
Maintenance efficacy designs in psychiatry: Randomized discontinuation trials - enriched but not better. J Clin Transl Sci 2017; 1:198-204. [PMID: 29082033 PMCID: PMC5647671 DOI: 10.1017/cts.2017.2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 01/26/2017] [Indexed: 12/17/2022] Open
Abstract
Introduction Although classical randomized clinical trials (RCTs) are the gold standard for proof of drug efficacy, randomized discontinuation trials (RDTs), sometimes called “enriched” trials, are used increasingly, especially in psychiatric maintenance studies. Methods A narrative review of two decades of experience with RDTs. Results RDTs in psychiatric maintenance trials tend to use a dependent variable as a predictor: treatment response. Treatment responders are assessed for treatment response. This tautology in the logic of RDTs renders them invalid, since the predictor and the outcome are the same variable. Although RDTs can be designed to avoid this tautologous state of affairs, like using independent predictors of outcomes, such is not the case with psychiatric maintenance studies Further, purported benefits of RDTs regarding feasibility were found to be questionable. Specifically, RDTs do not enhance statistical power in many settings, and, because of high dropout rates, produce results of questionable validity. Any claimed benefits come with notably reduced generalizability. Conclusions RDTs appear to be scientifically invalid as used in psychiatric maintenance designs. Their purported feasibility benefits are not seen in actual trials for psychotropic drugs. There is warrant for changes in federal policy regarding marketing indications for maintenance efficacy using the RDT design.
Collapse
|
12
|
Grossman SA, Schreck KC, Ballman K, Alexander B. Point/counterpoint: randomized versus single-arm phase II clinical trials for patients with newly diagnosed glioblastoma. Neuro Oncol 2017; 19:469-474. [PMID: 28388713 PMCID: PMC5464324 DOI: 10.1093/neuonc/nox030] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Stuart A Grossman
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, USA
| | - Karisa C Schreck
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, USA
| | | | | |
Collapse
|
13
|
de Jong EEC, van Elmpt W, Leijenaar RTH, Hoekstra OS, Groen HJM, Smit EF, Boellaard R, van der Noort V, Troost EGC, Lambin P, Dingemans AMC. [18F]FDG PET/CT-based response assessment of stage IV non-small cell lung cancer treated with paclitaxel-carboplatin-bevacizumab with or without nitroglycerin patches. Eur J Nucl Med Mol Imaging 2016; 44:8-16. [PMID: 27600280 PMCID: PMC5121177 DOI: 10.1007/s00259-016-3498-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/16/2016] [Indexed: 12/30/2022]
Abstract
PURPOSE Nitroglycerin (NTG) is a vasodilating drug, which increases tumor blood flow and consequently decreases hypoxia. Therefore, changes in [18F] fluorodeoxyglucose positron emission tomography ([18F]FDG PET) uptake pattern may occur. In this analysis, we investigated the feasibility of [18F]FDG PET for response assessment to paclitaxel-carboplatin-bevacizumab (PCB) treatment with and without NTG patches. And we compared the [18F]FDG PET response assessment to RECIST response assessment and survival. METHODS A total of 223 stage IV non-small cell lung cancer (NSCLC) patients were included in a phase II study (NCT01171170) randomizing between PCB treatment with or without NTG patches. For 60 participating patients, a baseline and a second [18F]FDG PET/computed tomography (CT) scan, performed between day 22 and 24 after the start of treatment, were available. Tumor response was defined as a 30 % decrease in CT and PET parameters, and was compared to RECIST response at week 6. The predictive value of these assessments for progression free survival (PFS) and overall survival (OS) was assessed with and without NTG. RESULTS A 30 % decrease in SUVpeak assessment identified more patients as responders compared to a 30 % decrease in CT diameter assessment (73 % vs. 18 %), however, this was not correlated to OS (SUVpeak30 p = 0.833; CTdiameter30 p = 0.557). Changes in PET parameters between the baseline and the second scan were not significantly different for the NTG group compared to the control group (p value range 0.159-0.634). The CT-based (part of the [18F]FDG PET/CT) parameters showed a significant difference between the baseline and the second scan for the NTG group compared to the control group (CT diameter decrease of 7 ± 23 % vs. 19 ± 14 %, p = 0.016, respectively). CONCLUSIONS The decrease in tumoral FDG uptake in advanced NSCLC patients treated with chemotherapy with and without NTG did not differ between both treatment arms. Early PET-based response assessment showed more tumor responders than CT-based response assessment (part of the [18F]FDG PET/CT); this was not correlated to survival. This might be due to timing of the [18F]FDG PET shortly after the bevacizumab infusion.
Collapse
Affiliation(s)
- Evelyn E C de Jong
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands.
| | - Wouter van Elmpt
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Ralph T H Leijenaar
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Otto S Hoekstra
- Department of Nuclear Medicine & PET Research, VU University Medical Center, Amsterdam, Netherlands
| | - Harry J M Groen
- Department of Pulmonary Diseases, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Egbert F Smit
- Department of Pulmonary Diseases, VU University Medical Center, Amsterdam, Netherlands
- Department of Thoracic Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, Netherlands
| | - Vincent van der Noort
- Department of Biometrics, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | - Esther G C Troost
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
- Institute of Radiooncology, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Medical Faculty and University Hospital Carl Gustav Carus of Technische Universität Dresden, Dresden, Germany
| | - Philippe Lambin
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Anne-Marie C Dingemans
- Department of Pulmonology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
14
|
Systematic review of enriched enrolment, randomised withdrawal trial designs in chronic pain: a new framework for design and reporting. Pain 2016; 156:1382-1395. [PMID: 25985142 DOI: 10.1097/j.pain.0000000000000088] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Enriched enrolment, randomised withdrawal (EERW) pain trials select, before randomisation, patients who respond by demonstrating a predetermined degree of pain relief and acceptance of adverse events. There is uncertainty over the value of this design. We report a systematic review of EERW trials in chronic noncancer pain together with a critical appraisal of methods and potential biases in the methods used and recommendations for the design and reporting of future EERW trials. Electronic and other searches found 25 EERW trials published between 1995 and June 2014, involving 5669 patients in a randomised withdrawal phase comparing drug with placebo; 13 (median, 107 patients) had a randomised withdrawal phase of 6 weeks or less, and 12 (median, 334) lasted 12 to 26 weeks. Risks of bias included short duration, inadequate outcome definition, incomplete outcome data reporting, small size, and inadequate dose tapering on randomisation to placebo. Active treatment was usually better than placebo (22/25 trials). This review reduces the uncertainty around the value of EERW trials in pain. If properly designed, conducted, and reported, they are feasible and useful for making decisions about pain therapies. Shorter, small studies can be explanatory; longer, larger studies can inform practice. Current evidence is inadequate for valid comparisons in outcome between EERW and classical trials, although no gross differences were found. This systematic review provides a framework for assessing potential biases and the value of the EERW trials, and for the design of future studies by making recommendations for the conduct and reporting of EERW trials.
Collapse
|
15
|
Barrett JE. Information-adaptive clinical trials: a selective recruitment design. J R Stat Soc Ser C Appl Stat 2016. [DOI: 10.1111/rssc.12146] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
16
|
Marconato L, Buracco P, Aresu L. Perspectives on the design of clinical trials for targeted therapies and immunotherapy in veterinary oncology. Vet J 2015; 205:238-43. [DOI: 10.1016/j.tvjl.2015.02.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 02/15/2015] [Accepted: 02/25/2015] [Indexed: 12/18/2022]
|
17
|
|
18
|
Ivanova A, Rosner GL, Marchenko O, Parke T, Perevozskaya I, Wang Y. Advances in Statistical Approaches Oncology Drug Development. Ther Innov Regul Sci 2014; 48:81-89. [PMID: 25949927 DOI: 10.1177/2168479013501309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
We describe some recent developments in statistical methodology and practice in oncology drug development from an academic and an industry perspective. Many adaptive designs were pioneered in oncology, and oncology is still at the forefront of novel methods to enable better and faster Go/No-Go decision making while controlling the cost.
Collapse
Affiliation(s)
- Anastasia Ivanova
- Department of Biostatistics, University of North Carolina at Chapel Hill, NC, USA
| | - Gary L Rosner
- Oncology Biostatistics & Bioinformatics, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | | | - Tom Parke
- Tessella, Abingdon, Oxfordshire, England
| | - Inna Perevozskaya
- Statistical Research and Consulting Center, Pfizer, Inc., Collegeville, PA, USA
| | - Yanping Wang
- Biometrics and Advanced Analytics, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
19
|
Veterinary oncology clinical trials: design and implementation. Vet J 2014; 205:226-32. [PMID: 25582798 DOI: 10.1016/j.tvjl.2014.12.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 11/20/2014] [Accepted: 12/14/2014] [Indexed: 11/20/2022]
Abstract
There has been a recent increase in interest among veterinarians and the larger biomedical community in the evaluation of novel cancer therapies in client-owned (pet) animals with spontaneous cancer. This includes novel drugs designed to be veterinary therapeutics, as well as agents for which data generated in animals with tumors may inform human clinical trial design and implementation. An understanding of the process involved in moving a therapeutic agent through the stages of clinical evaluation is critical to the successful implementation of clinical investigations, as well as interpretation of the veterinary oncology literature. This review outlines considerations in the design and conduct of the various phases of oncology clinical trials, along with recent adaptations/modifications of these basic designs that can enhance the generation of timely and meaningful clinical data.
Collapse
|
20
|
Chuang-Stein C, Follman D, Chappell R. University of Pennsylvania 6th annual conference on statistical issues in clinical trials: Dynamic treatment regimes (afternoon session). Clin Trials 2014; 11:457-466. [PMID: 25053777 DOI: 10.1177/1740774514538552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
21
|
|
22
|
Papadimitrakopoulou VA. Novel Clinical Trial Designs for Metastatic Lung Cancer. Lung Cancer 2014. [DOI: 10.1002/9781118468791.ch31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
23
|
Fedorov VV, Liu T. Randomized Discontinuation Trials With Binary Outcomes. JOURNAL OF STATISTICAL THEORY AND PRACTICE 2014. [DOI: 10.1080/15598608.2014.840492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
24
|
Ledermann JA, Ray-Coquard I. Novel approaches to improve the treatment of rare gynecologic cancers: research opportunities and challenges. Am Soc Clin Oncol Educ Book 2014:e282-e286. [PMID: 24857114 DOI: 10.14694/edbook_am.2014.34.e282] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
More than 50% of all gynecologic cancers can be classified as rare tumors (defined as an incidence of fewer than six per 100,000). Improved understanding of the molecular pathogenesis of tumors increases the proportion of rare tumors and creates challenges in optimizing the design of clinical trials. Novel trial designs are needed to take forward the development of new treatments in rare tumors. This requires international partnerships, harmonization of treatment, and collaboration to overcome the regulatory barriers to conducting international trials. Although randomized trials can be done in many tumor types, there are some for which conducting even single-arm studies may be challenging. For these tumors, robust collection of data through national and/or international registries could lead through audit to improvements in the treatment of rare tumors.
Collapse
Affiliation(s)
- Jonathan A Ledermann
- From the UCL Cancer Institute, London, United Kingdom; Department of Adult Medical Oncology, Centre Leon Berard, Lyon, France
| | - Isabelle Ray-Coquard
- From the UCL Cancer Institute, London, United Kingdom; Department of Adult Medical Oncology, Centre Leon Berard, Lyon, France
| |
Collapse
|
25
|
Qin R, Kohli M. Pharmacogenetics- and pharmacogenomics-based rational clinical trial designs in oncology. Per Med 2013; 10:859-869. [PMID: 29776282 DOI: 10.2217/pme.13.78] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The rapid evolution of molecular technologies that can identify genetic markers and lead to dissecting the inherent variance of individual cancer biology has had a tangible impact on trial designs in oncology. Rational trial designs based on molecular marker expression coupled with drug–marker interactions have started to be adopted, challenging the previous paradigms of morphology-based, single-arm efficacy studies. This review summarizes novel trials being developed based on molecular predictive factor therapeutics and the potential impact these novel trial designs will have on the practice of oncology in future. A variety of clinical trial designs based on tumor and drug–host genetic interactions are discussed and the example of advanced prostate cancer is used to illustrate the changing landscape of clinical trial designs in cancer.
Collapse
Affiliation(s)
- Rui Qin
- Department of Health Sciences Research, Mayo Clinic, 200 First Street South West, Rochester, MN 55905, USA
| | - Manish Kohli
- Department of Oncology, Mayo Clinic, 200 First Street South West, Rochester, MN 55905, USA
| |
Collapse
|
26
|
Hong F, Simon R. Run-in phase III trial design with pharmacodynamics predictive biomarkers. J Natl Cancer Inst 2013; 105:1628-33. [PMID: 24096624 DOI: 10.1093/jnci/djt265] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Developments in biotechnology have stimulated the use of predictive biomarkers to identify patients who are likely to benefit from a targeted therapy. Several randomized phase III designs have been introduced for development of a targeted therapy using a diagnostic test. Most such designs require biomarkers measured before treatment. In many cases, it has been very difficult to identify such biomarkers. Promising candidate biomarkers can sometimes be effectively measured after a short run-in period on the new treatment. METHODS We introduce a new design for phase III trials with a candidate predictive pharmacodynamic biomarker measured after a short run-in period. Depending on the therapy and the biomarker performance, the trial would either randomize all patients but perform a separate analysis on the biomarker-positive patients or only randomize marker-positive patients after the run-in period. We evaluate the proposed design compared with the conventional phase III design and discuss how to design a run-in trial based on phase II studies. RESULTS The proposed design achieves a major sample size reduction compared with the conventional randomized phase III design in many cases when the biomarker has good sensitivity (≥0.7) and specificity (≥0.7). This requires that the biomarker be measured accurately and be indicative of drug activity. However, the proposed design loses some of its advantage when the proportion of potential responders is large (>50%) or the effect on survival from run-in period is substantial. CONCLUSIONS Incorporating a pharmacodynamic biomarker requires careful consideration but can expand the capacity of clinical trials to personalize treatment decisions and enhance therapeutics development.
Collapse
Affiliation(s)
- Fangxin Hong
- Affiliations of authors: Departments of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard School of Public Health, Boston, MA (FH); Biometric Research Branch, National Cancer Institute, Bethesda, MD (RS)
| | | |
Collapse
|
27
|
Teng FF, Meng X, Sun XD, Yu JM. New strategy for monitoring targeted therapy: molecular imaging. Int J Nanomedicine 2013; 8:3703-13. [PMID: 24124361 PMCID: PMC3794840 DOI: 10.2147/ijn.s51264] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Targeted therapy is becoming an increasingly important component in the treatment of cancer. How to accurately monitor targeted therapy has been crucial in clinical practice. The traditional approach to monitor treatment through imaging has relied on assessing the change of tumor size by refined World Health Organization criteria, or more recently, by the Response Evaluation Criteria in Solid Tumors. However, these criteria, which are based on the change of tumor size, show some limitations for evaluating targeted therapy. Currently, genetic alterations are identified with prognostic as well as predictive potential concerning the use of molecularly targeted drugs. Conversely, considering the limitations of invasiveness and the issue of expression heterogeneity, molecular imaging is better able to assay in vivo biologic processes noninvasively and quantitatively, and has been a particularly attractive tool for monitoring treatment in clinical cancer practice. This review focuses on the applications of different kinds of molecular imaging including positron emission tomography-, magnetic resonance imaging-, ultrasonography-, and computed tomography-based imaging strategies on monitoring targeted therapy. In addition, the key challenges of molecular imaging are addressed to successfully translate these promising techniques in the future.
Collapse
Affiliation(s)
- Fei-Fei Teng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong University, Jinan, People's Republic of China
| | | | | | | |
Collapse
|
28
|
Chevreau C, Le Cesne A, Ray-Coquard I, Italiano A, Cioffi A, Isambert N, Robin YM, Fournier C, Clisant S, Chaigneau L, Bay JO, Bompas E, Gauthier E, Blay JY, Penel N. Sorafenib in patients with progressive epithelioid hemangioendothelioma: a phase 2 study by the French Sarcoma Group (GSF/GETO). Cancer 2013; 119:2639-44. [PMID: 23589078 DOI: 10.1002/cncr.28109] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 01/24/2013] [Accepted: 01/25/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND There is no standard treatment for progressive epithelioid hemangioendothelioma (EHE). To investigate the significant vascularization of EHE, the activity/toxicity of sorafenib in patients with progressive EHE was explored. METHODS In this multicenter, 1-stage, phase 2 trial of sorafenib (800 mg daily), the primary endpoint, which was chosen by default, was the 9-month progression-free rate. All patients had documented progressive disease at the time of study entry. RESULTS Fifteen patients were enrolled between June 2009 and February 2011. The median age was 57 years (range, 31-76 years), and the ratio of men to women was 9:6. The performance status was zero in 10 patients and 1 in 5 patients. Twelve patients had metastases, mainly in the lung (12 patients), liver (5 patients), and bone (3 patients). Five patients had received prior chemotherapy (doxorubicin in 5 patients and taxane in 3 patients). The median sorafenib treatment duration was 124 days (range, from 27 to >271 days). Seven patients required dose reductions or transient treatment discontinuation. The 9-month progression-free rate was 30.7% (4 of 13 patients). The 2-month, 4-month, and 6-month progression-free rate was 84.6% (11 of 13 patients), 46.4% (6 of 13 patients), and 38.4% (5 of 13 patients), respectively. Two partial responses were observed that lasted 2 months and 9 months. CONCLUSIONS Further clinical trials exploring sorafenib as treatment of progressive EHE are needed.
Collapse
Affiliation(s)
- Christine Chevreau
- Department of Medical Oncology, Claudius Regaud Institute, Toulouse, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Karashima T, Fukuhara H, Tamura K, Ashida S, Kamada M, Inoue K, Taguchi T, Kuroda N, Shuin T. Expression of angiogenesis-related gene profiles and development of resistance to tyrosine-kinase inhibitor in advanced renal cell carcinoma: Characterization of sorafenib-resistant cells derived from a cutaneous metastasis. Int J Urol 2013; 20:923-30. [DOI: 10.1111/iju.12084] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 12/19/2012] [Indexed: 12/19/2022]
Affiliation(s)
| | - Hideo Fukuhara
- Department of Urology; Kochi University; Nankoku City; Japan
| | - Kenji Tamura
- Department of Urology; Kochi University; Nankoku City; Japan
| | - Shingo Ashida
- Department of Urology; Kochi University; Nankoku City; Japan
| | - Masayuki Kamada
- Department of Urology; Kochi University; Nankoku City; Japan
| | - Keiji Inoue
- Department of Urology; Kochi University; Nankoku City; Japan
| | - Takahiro Taguchi
- Department of Human Health and Medical Science; Faculty of Medicine; Kochi University; Nankoku City; Japan
| | - Naoto Kuroda
- Department of Pathology; Kochi Red Cross Hospital; Kochi City; Japan
| | - Taro Shuin
- Department of Urology; Kochi University; Nankoku City; Japan
| |
Collapse
|
30
|
Karrison TG, Ratain MJ, Stadler WM, Rosner GL. Estimation of Progression-Free Survival for All Treated Patients in the Randomized Discontinuation Trial Design. AM STAT 2012; 66:155-162. [PMID: 24039273 DOI: 10.1080/00031305.2012.720900] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The randomized discontinuation trial (RDT) design is an enrichment-type design that has been used in a variety of diseases to evaluate the efficacy of new treatments. The RDT design seeks to select a more homogeneous group of patients, consisting of those who are more likely to show a treatment benefit if one exists. In oncology, the RDT design has been applied to evaluate the effects of cytostatic agents, that is, drugs that act primarily by slowing tumor growth rather than shrinking tumors. In the RDT design, all patients receive treatment during an initial, open-label run-in period of duration T. Patients with objective response (substantial tumor shrinkage) remain on therapy while those with early progressive disease are removed from the trial. Patients with stable disease (SD) are then randomized to either continue active treatment or switched to placebo. The main analysis compares outcomes, for example, progression-free survival (PFS), between the two randomized arms. As a secondary objective, investigators may seek to estimate PFS for all treated patients, measured from the time of entry into the study, by combining information from the run-in and post run-in periods. For t ≤ T, PFS is estimated by the observed proportion of patients who are progression-free among all patients enrolled. For t > T, the estimate can be expressed as Ŝ(t) = p̂OR × ŜOR(t - T) + p̂SD × ŜSD(t - T), where p̂OR is the estimated probability of response during the run-in period, p̂SD is the estimated probability of SD, and ŜOR(t - T) and ŜSD(t - T) are the Kaplan-Meier estimates of subsequent PFS in the responders and patients with SD randomized to continue treatment, respectively. In this article, we derive the variance of Ŝ(t), enabling the construction of confidence intervals for both S(t) and the median survival time. Simulation results indicate that the method provides accurate coverage rates. An interesting aspect of the design is that outcomes during the run-in phase have a negative multinomial distribution, something not frequently encountered in practice.
Collapse
Affiliation(s)
- Theodore G Karrison
- Department of Health Studies, University of Chicago, 5841 S. Maryland Ave., MC2007, Chicago, IL 60637
| | | | | | | |
Collapse
|
31
|
|
32
|
Early phase drug development for treatment of chronic pain — Options for clinical trial and program design. Contemp Clin Trials 2012; 33:689-99. [DOI: 10.1016/j.cct.2012.02.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 02/20/2012] [Accepted: 02/22/2012] [Indexed: 11/22/2022]
|
33
|
Galanis E, Wu W, Cloughesy T, Lamborn K, Mann B, Wen PY, Reardon DA, Wick W, Macdonald D, Armstrong TS, Weller M, Vogelbaum M, Colman H, Sargent DJ, van den Bent MJ, Gilbert M, Chang S. Phase 2 trial design in neuro-oncology revisited: a report from the RANO group. Lancet Oncol 2012; 13:e196-204. [PMID: 22554547 DOI: 10.1016/s1470-2045(11)70406-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Advances in the management of gliomas, including the approval of agents such as temozolomide and bevacizumab, have created an evolving therapeutic landscape in glioma treatment, thus affecting our ability to reliably use historical controls to comparatively assess the activity of new therapies. Furthermore, the increasing availability of novel, targeted agents--which are competing for a small patient population, in view of the low incidence of primary brain tumours--draws attention to the need to improve the efficiency of phase 2 clinical testing in neuro-oncology to expeditiously transition the most promising of these drugs or combinations to potentially practice-changing phase 3 trials. In this report from the Response Assessment in Neurooncology (RANO) group, we review phase 2 trial designs that can address these challenges and capitalise on scientific and clinical advances in brain tumour treatment in neuro-oncology to accelerate and optimise the selection of drugs deserving further testing in phase 3 trials. Although there is still a small role for single-arm and non-comparative phase 2 designs, emphasis is placed on the potential role that comparative randomised phase 2 designs--such as screening designs, selection designs, discontinuation designs, and adaptive designs, including seamless phase 2/3 designs--can have. The rational incorporation of these designs, as determined by the specific clinical setting and the trial's endpoints or goals, has the potential to substantially advance new drug development in neuro-oncology.
Collapse
|
34
|
Kang H, Lee HY, Lee KS, Kim JH. Imaging-based tumor treatment response evaluation: review of conventional, new, and emerging concepts. Korean J Radiol 2012; 13:371-90. [PMID: 22778559 PMCID: PMC3384819 DOI: 10.3348/kjr.2012.13.4.371] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 05/14/2012] [Indexed: 01/07/2023] Open
Abstract
Tumor response may be assessed readily by the use of Response Evaluation Criteria in Solid Tumor version 1.1. However, the criteria mainly depend on tumor size changes. These criteria do not reflect other morphologic (tumor necrosis, hemorrhage, and cavitation), functional, or metabolic changes that may occur with targeted chemotherapy or even with conventional chemotherapy. The state-of-the-art multidetector CT is still playing an important role, by showing high-quality, high-resolution images that are appropriate enough to measure tumor size and its changes. Additional imaging biomarker devices such as dual energy CT, positron emission tomography, MRI including diffusion-weighted MRI shall be more frequently used for tumor response evaluation, because they provide detailed anatomic, and functional or metabolic change information during tumor treatment, particularly during targeted chemotherapy. This review elucidates morphologic and functional or metabolic approaches, and new concepts in the evaluation of tumor response in the era of personalized medicine (targeted chemotherapy).
Collapse
Affiliation(s)
- Hee Kang
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Korea
| | | | | | | |
Collapse
|
35
|
Yanagawa M, Tatsumi M, Miyata H, Morii E, Tomiyama N, Watabe T, Isohashi K, Kato H, Shimosegawa E, Yamasaki M, Mori M, Doki Y, Hatazawa J. Evaluation of response to neoadjuvant chemotherapy for esophageal cancer: PET response criteria in solid tumors versus response evaluation criteria in solid tumors. J Nucl Med 2012; 53:872-80. [PMID: 22582049 DOI: 10.2967/jnumed.111.098699] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Recently, PET response criteria in solid tumors (PERCIST) have been proposed as a new standardized method to assess chemotherapeutic response metabolically and quantitatively. The aim of this study was to evaluate therapeutic response to neoadjuvant chemotherapy for locally advanced esophageal cancer, comparing PERCIST with the currently widely used response evaluation criteria in solid tumors (RECIST). METHODS Fifty-one patients with locally advanced esophageal cancer who received neoadjuvant chemotherapy (5-fluorouracil, adriamycin, and cisplatin), followed by surgery were studied. Chemotherapeutic lesion responses were evaluated using (18)F-FDG PET and CT according to the RECIST and PERCIST methods. The PET/CT scans were obtained before chemotherapy and about 2 wk after completion of chemotherapy. Associations were statistically analyzed between survival (overall and disease-free survival) and clinicopathologic results (histology [well-, moderately, and poorly differentiated squamous cell carcinoma], lymphatic invasion, venous invasion, clinical stage, pathologic stage, resection level, reduction rate of tumor diameter, reduction rate of tumor uptake, chemotherapeutic responses in RECIST and PERCIST, and pathologic response). RESULTS There was a significant difference in response classification between RECIST and PERCIST (Wilcoxon signed-rank test, P < 0.0001). Univariate analysis showed that lymphatic invasion, venous invasion, resection level, pathologic stage, and PERCIST were significant factors associated with disease-free or overall survival in this study. Although multivariate analysis demonstrated that venous invasion (disease-free survival: hazard ratio [HR] = 4.519, P = 0.002; overall survival: HR = 5.591, P = 0.003) and resection level (disease-free survival: HR = 11.078, P = 0.001) were the significant predictors, PERCIST was also significant in noninvasive therapy response assessment before surgery (disease-free survival: HR = 4.060, P = 0.025; overall survival: HR = 8.953, P = 0.034). CONCLUSION RECIST based on the anatomic size reduction rate did not demonstrate the correlation between therapeutic responses and prognosis in patients with esophageal cancer receiving neoadjuvant chemotherapy. However, PERCIST was found to be the strongest independent predictor of outcomes. Given the significance of noninvasive radiologic imaging in formulating clinical treatment strategies, PERCIST might be considered more suitable for evaluation of chemotherapeutic response to esophageal cancer than RECIST.
Collapse
Affiliation(s)
- Masahiro Yanagawa
- Department of Radiology, Osaka University Graduate School of Medicine, Suita-city, Osaka, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Nosov DA, Esteves B, Lipatov ON, Lyulko AA, Anischenko AA, Chacko RT, Doval DC, Strahs A, Slichenmyer WJ, Bhargava P. Antitumor Activity and Safety of Tivozanib (AV-951) in a Phase II Randomized Discontinuation Trial in Patients With Renal Cell Carcinoma. J Clin Oncol 2012; 30:1678-85. [DOI: 10.1200/jco.2011.35.3524] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose The antitumor activity and safety of tivozanib, which is a potent and selective vascular endothelial growth factor receptor-1, -2, and -3 inhibitor, was assessed in patients with advanced/metastatic renal cell carcinoma (RCC). Patients and Methods In this phase II, randomized discontinuation trial, 272 patients received open-label tivozanib 1.5 mg/d (one cycle equaled three treatment weeks followed by a 1-week break) orally for 16 weeks. Thereafter, 78 patients who demonstrated ≥ 25% tumor shrinkage continued to take tivozanib, and 118 patients with less than 25% tumor change were randomly assigned to receive tivozanib or a placebo in a double-blind manner; patients with ≥ 25% tumor growth were discontinued. Primary end points included safety, the objective response rate (ORR) at 16 weeks, and the percentage of randomly assigned patients who remained progression free after 12 weeks of double-blind treatment; secondary end points included progression-free survival (PFS). Results Of 272 patients enrolled onto the study, 83% of patients had clear-cell histology, 73% of patients had undergone nephrectomy, and 54% of patients were treatment naive. The ORR after 16 weeks of tivozanib treatment was 18% (95% CI, 14% to 23%). Of the 118 randomized patients, significantly more patients who were randomly assigned to receive double-blind tivozanib remained progression free after 12 weeks versus patients who received the placebo (49% v 21%; P = .001). Throughout the study, the ORR was 24% (95% CI, 19% to 30%), and the median PFS was 11.7 months (95% CI, 8.3 to 14.3 months) in the overall study population. The most common grade 3 and 4 treatment-related adverse event was hypertension (12%). Conclusion Tivozanib was active and well tolerated in patients with advanced RCC. These data support additional development of tivozanib in advanced RCC.
Collapse
Affiliation(s)
- Dmitry A. Nosov
- Dmitry A. Nosov, Blokhin Cancer Research Center, Moscow; Oleg N. Lipatov, Bashkortostan Clinical Oncology Center, Ufa, Russia; Brooke Esteves, Andrew Strahs, William J. Slichenmyer, and Pankaj Bhargava, AVEO Pharmaceuticals, Cambridge; Pankaj Bhargava, Dana-Farber Cancer Institute, Boston, MA; Alexei A. Lyulko, Zaporizhya Medical Academy of Postgraduate Education, Zaporizhya; A.A. Anischenko, Donetsk Regional Antitumor Center, Donetsk, Ukraine; Raju T. Chacko, Christian Medical College, Vellore; and
| | - Brooke Esteves
- Dmitry A. Nosov, Blokhin Cancer Research Center, Moscow; Oleg N. Lipatov, Bashkortostan Clinical Oncology Center, Ufa, Russia; Brooke Esteves, Andrew Strahs, William J. Slichenmyer, and Pankaj Bhargava, AVEO Pharmaceuticals, Cambridge; Pankaj Bhargava, Dana-Farber Cancer Institute, Boston, MA; Alexei A. Lyulko, Zaporizhya Medical Academy of Postgraduate Education, Zaporizhya; A.A. Anischenko, Donetsk Regional Antitumor Center, Donetsk, Ukraine; Raju T. Chacko, Christian Medical College, Vellore; and
| | - Oleg N. Lipatov
- Dmitry A. Nosov, Blokhin Cancer Research Center, Moscow; Oleg N. Lipatov, Bashkortostan Clinical Oncology Center, Ufa, Russia; Brooke Esteves, Andrew Strahs, William J. Slichenmyer, and Pankaj Bhargava, AVEO Pharmaceuticals, Cambridge; Pankaj Bhargava, Dana-Farber Cancer Institute, Boston, MA; Alexei A. Lyulko, Zaporizhya Medical Academy of Postgraduate Education, Zaporizhya; A.A. Anischenko, Donetsk Regional Antitumor Center, Donetsk, Ukraine; Raju T. Chacko, Christian Medical College, Vellore; and
| | - Alexei A. Lyulko
- Dmitry A. Nosov, Blokhin Cancer Research Center, Moscow; Oleg N. Lipatov, Bashkortostan Clinical Oncology Center, Ufa, Russia; Brooke Esteves, Andrew Strahs, William J. Slichenmyer, and Pankaj Bhargava, AVEO Pharmaceuticals, Cambridge; Pankaj Bhargava, Dana-Farber Cancer Institute, Boston, MA; Alexei A. Lyulko, Zaporizhya Medical Academy of Postgraduate Education, Zaporizhya; A.A. Anischenko, Donetsk Regional Antitumor Center, Donetsk, Ukraine; Raju T. Chacko, Christian Medical College, Vellore; and
| | - A. A. Anischenko
- Dmitry A. Nosov, Blokhin Cancer Research Center, Moscow; Oleg N. Lipatov, Bashkortostan Clinical Oncology Center, Ufa, Russia; Brooke Esteves, Andrew Strahs, William J. Slichenmyer, and Pankaj Bhargava, AVEO Pharmaceuticals, Cambridge; Pankaj Bhargava, Dana-Farber Cancer Institute, Boston, MA; Alexei A. Lyulko, Zaporizhya Medical Academy of Postgraduate Education, Zaporizhya; A.A. Anischenko, Donetsk Regional Antitumor Center, Donetsk, Ukraine; Raju T. Chacko, Christian Medical College, Vellore; and
| | - Raju T. Chacko
- Dmitry A. Nosov, Blokhin Cancer Research Center, Moscow; Oleg N. Lipatov, Bashkortostan Clinical Oncology Center, Ufa, Russia; Brooke Esteves, Andrew Strahs, William J. Slichenmyer, and Pankaj Bhargava, AVEO Pharmaceuticals, Cambridge; Pankaj Bhargava, Dana-Farber Cancer Institute, Boston, MA; Alexei A. Lyulko, Zaporizhya Medical Academy of Postgraduate Education, Zaporizhya; A.A. Anischenko, Donetsk Regional Antitumor Center, Donetsk, Ukraine; Raju T. Chacko, Christian Medical College, Vellore; and
| | - Dinesh C. Doval
- Dmitry A. Nosov, Blokhin Cancer Research Center, Moscow; Oleg N. Lipatov, Bashkortostan Clinical Oncology Center, Ufa, Russia; Brooke Esteves, Andrew Strahs, William J. Slichenmyer, and Pankaj Bhargava, AVEO Pharmaceuticals, Cambridge; Pankaj Bhargava, Dana-Farber Cancer Institute, Boston, MA; Alexei A. Lyulko, Zaporizhya Medical Academy of Postgraduate Education, Zaporizhya; A.A. Anischenko, Donetsk Regional Antitumor Center, Donetsk, Ukraine; Raju T. Chacko, Christian Medical College, Vellore; and
| | - Andrew Strahs
- Dmitry A. Nosov, Blokhin Cancer Research Center, Moscow; Oleg N. Lipatov, Bashkortostan Clinical Oncology Center, Ufa, Russia; Brooke Esteves, Andrew Strahs, William J. Slichenmyer, and Pankaj Bhargava, AVEO Pharmaceuticals, Cambridge; Pankaj Bhargava, Dana-Farber Cancer Institute, Boston, MA; Alexei A. Lyulko, Zaporizhya Medical Academy of Postgraduate Education, Zaporizhya; A.A. Anischenko, Donetsk Regional Antitumor Center, Donetsk, Ukraine; Raju T. Chacko, Christian Medical College, Vellore; and
| | - William J. Slichenmyer
- Dmitry A. Nosov, Blokhin Cancer Research Center, Moscow; Oleg N. Lipatov, Bashkortostan Clinical Oncology Center, Ufa, Russia; Brooke Esteves, Andrew Strahs, William J. Slichenmyer, and Pankaj Bhargava, AVEO Pharmaceuticals, Cambridge; Pankaj Bhargava, Dana-Farber Cancer Institute, Boston, MA; Alexei A. Lyulko, Zaporizhya Medical Academy of Postgraduate Education, Zaporizhya; A.A. Anischenko, Donetsk Regional Antitumor Center, Donetsk, Ukraine; Raju T. Chacko, Christian Medical College, Vellore; and
| | - Pankaj Bhargava
- Dmitry A. Nosov, Blokhin Cancer Research Center, Moscow; Oleg N. Lipatov, Bashkortostan Clinical Oncology Center, Ufa, Russia; Brooke Esteves, Andrew Strahs, William J. Slichenmyer, and Pankaj Bhargava, AVEO Pharmaceuticals, Cambridge; Pankaj Bhargava, Dana-Farber Cancer Institute, Boston, MA; Alexei A. Lyulko, Zaporizhya Medical Academy of Postgraduate Education, Zaporizhya; A.A. Anischenko, Donetsk Regional Antitumor Center, Donetsk, Ukraine; Raju T. Chacko, Christian Medical College, Vellore; and
| |
Collapse
|
37
|
Sleijfer S, Wagner AJ. The Challenge of Choosing Appropriate End Points in Single-Arm Phase II Studies of Rare Diseases. J Clin Oncol 2012; 30:896-8. [DOI: 10.1200/jco.2011.40.6942] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Stefan Sleijfer
- Erasmus University Medical Center, Daniel den Hoed Cancer Center, Rotterdam, the Netherlands
| | - Andrew J. Wagner
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
38
|
Gönen M. Discussions. Biometrics 2012; 68:215-6; discussion 224-5. [DOI: 10.1111/j.1541-0420.2011.01625.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
39
|
Perevozskaya I. Discussions. Biometrics 2012; 68:217-8; discussion 224-5. [DOI: 10.1111/j.1541-0420.2011.01626.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
40
|
Larkin JMG, Kipps ELS, Powell CJ, Swanton C. Systemic therapy for advanced renal cell carcinoma. Ther Adv Med Oncol 2011; 1:15-27. [PMID: 21789110 DOI: 10.1177/1758834009338430] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Renal cell carcinoma (RCC) accounts for approximately 3% of all cancers and is refractory to cytotoxic chemotherapy - immunotherapy has until recently been the standard of care for advanced disease. Randomised trials reported in the last 5 years have demonstrated that a number of agents including the monoclonal antibody, bevacizumab, and the kinase inhibitors - sorafenib sunitinib, temsirolimus and everolimus - are active in advanced RCC. Bevacizumab is directed against the vascular endothelial growth factor (VEGF), a key mediator of angiogenesis, whilst sorafenib and sunitinib inhibit a number of targets including the VEGF and platelet-derived growth factor (PDGFR) receptor tyrosine kinases. Temsirolimus and everolimus inhibit the intracellular mammalian target of rapamycin (mTOR) kinase. Sunitinib and temsirolimus have demonstrated efficacy in comparison with immunotherapy in the first-line setting in patients with favourable and poor prognosis advanced disease respectively. In the second-line setting, everolimus has shown benefit over placebo in patients who progress following treatment with a VEGF receptor tyrosine kinase inhibitor and sorafenib has demonstrated efficacy in comparison with placebo in patients with immunotherapy-refractory disease. We review here recent clinical trial data and discuss future developments in the systemic treatment of RCC including combination and sequential therapy, adjuvant therapy, the role of biomarkers and the prospects for the development of rational mechanism-directed therapy in this disease.
Collapse
Affiliation(s)
- James M G Larkin
- Department of Medicine, Royal Marsden Hospital, London SW3 6JJ, UK
| | | | | | | |
Collapse
|
41
|
Abstract
Renal cell cancer (RCC) has an increasing incidence internationally and is a disease for which there have been limited therapeutic options until recently. The last decade has seen a vastly improved understanding of the biological and clinical factors that predict the outcome of this disease. We now understand some of the different molecular underpinnings of renal clear cell carcinoma by mutation or silencing of the von Hippel Lindau (VHL) gene and subsequent deregulated proliferation and angiogenesis. Survival in advanced disease is predicted by factors (performance status, anemia, hypercalcemia, and serum lactate dehydrogenase, time from diagnosis to recurrence) incorporated into the Memorial Sloan Kettering Cancer Center (MSKCC) criteria (also referred to as 'Motzer' criteria). These criteria allow classification of patients with RCC into good, intermediate and poor risk categories with median overall survivals of 22 months, 12 months and 5.4 months, respectively. Predicated upon these advances, six new targeted drugs (sorafenib, sunitinib, temsirolimus, everolimus, bevacizumab and pazopanib) have been tested in well-designed phase III trials, selected or stratified for MSKCC risk criteria, with positive results. All of these new drugs act at least in part through vascular endothelial growth factor (VEGF) mediated pathways with other potential therapeutic impact on platelet-derived growth factor (PDGF), raf kinase and mammalian target of rapamycin (mTOR) pathways. Importantly, data from each of these trials show a consistent doubling of progression-free survival (PFS) over prior standard of care treatments. In addition, sorafenib, sunitinib and temsirolimus, have demonstrated significant overall survival (OS) benefits as well; further follow-up is required to determine whether the disease control exhibited by everolimus and pazopanib will translate into a survival advantage. These drugs are generally well tolerated, as demonstrated by quality-of-life improvement in clinical trials, and result in clinical benefit for in excess of 70% of patients treated. They have challenged the traditional outcomes of clinical trial design by achieving their benefits with relatively few radiographic responses, but high rates of disease stability. The unique side-effect profile coupled with the chronicity of therapy requires increased vigilance to maximize exposure to the drugs while maintaining quality of life and minimizing toxicity. This review focuses on the background, clinical development and practical use of these new drugs in RCC.
Collapse
Affiliation(s)
- Tanya B Dorff
- Assistant Professors of Medicine, Kenneth J. Norris Comprehensive Cancer Center, Section of Genitourinary Medical Oncology, Division of Cancer Medicine and Blood Diseases, University of Southern California, Los Angeles CA, USA
| | | | | |
Collapse
|
42
|
Gill S, Berry S, Biagi J, Butts C, Buyse M, Chen E, Jonker D, Mărginean C, Samson B, Stewart J, Thirlwell M, Wong R, Maroun J. Progression-free survival as a primary endpoint in clinical trials of metastatic colorectal cancer. Curr Oncol 2011; 18 Suppl 2:S5-S10. [PMID: 21969810 PMCID: PMC3176908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023] Open
Abstract
In recent years, significant advances have been made in the management of metastatic colorectal cancer. Traditionally, an improvement in overall survival has been considered the "gold standard"-the most convincing measure of efficacy. However, overall survival requires larger patient numbers and longer follow-up and may often be confounded by other factors, including subsequent therapies and crossover. Given the number of active therapies for potential investigation, demand for rapid evaluation and early availability of new therapies is growing. Progression-free survival is regarded as an important measure of treatment benefit and, compared with overall survival, can be evaluated earlier, with fewer patients and no confounding by subsequent lines of therapy. The present paper reviews the advantages, limitations, and relevance of progression-free survival as a primary endpoint in randomized trials of metastatic colorectal cancer.
Collapse
Affiliation(s)
- S. Gill
- University of British Columbia, Division of Medical Oncology, and BC Cancer Agency, Vancouver, BC
| | - S. Berry
- Sunnybrook Odette Cancer Centre and University of Toronto, Toronto, ON
| | - J. Biagi
- Department of Oncology, Queen’s University, Kingston, ON
| | - C. Butts
- Cross Cancer Institute and University of Alberta, Edmonton, AB
| | - M. Buyse
- International Drug Development Institute, Louvain-la-Neuve, and I-BioStat, Hasselt University, Diepenbeek, Belgium
| | - E. Chen
- Department of Medical Oncology and Hematology, Princess Margaret Hospital, Toronto, ON
| | - D. Jonker
- Ottawa Hospital Cancer Centre, Ottawa, ON
| | | | - B. Samson
- Centre intégré de cancer de la Montérégie de l’Hôpital Charles-Lemoyne, Greenfield Park, QC
| | - J. Stewart
- University Health Network, Princess Margaret Hospital, Toronto, ON
| | - M. Thirlwell
- McGill University, McGill University Health Centre, and Montreal General Hospital, Montreal, QC
| | - R. Wong
- CancerCare Manitoba, University of Manitoba, Winnipeg, MB
| | - J.A. Maroun
- Ottawa Hospital Cancer Centre and University of Ottawa, Ottawa, ON
| |
Collapse
|
43
|
Abstract
Classical phase II trial designs, including "adaptive" designs, require the prospective characterization of tumors. We propose a 2-stage phase II design that allows for characterization of tumors and selection of a tumor subtype of interest at the conclusion of stage 1. The stage 2 objective is either a classical estimate of the response rate for either the tumor or a subtype, or a formal test of the hypothesis that the response rate for a subtype is greater than the overall response rate. Considering likely scenarios, stage 1 sample sizes approximately range from 20 to 100 with a usual size of 50. This compares with typical classical stage 1 sample sizes of 12 to 30. Total sample sizes range from sizes identical to classical designs (tens to scores) to large sizes typical of phase III trials in metastatic disease (hundreds). Our design is more efficient than previous adaptive designs because it allows for the selection of a tumor subtype of interest on the basis of results from stage 1. It complements classical phase II and phase III designs in which investigators compare different treatments in similar patients and tumors by positioning a treatment as fixed (control) and using tumor subtype as the variable of interest.
Collapse
Affiliation(s)
- John D Roberts
- Massey Cancer Center and Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | | |
Collapse
|
44
|
Trippa L, Rosner GL, Müller P. Bayesian enrichment strategies for randomized discontinuation trials. Biometrics 2011; 68:203-11. [PMID: 21714780 DOI: 10.1111/j.1541-0420.2011.01623.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We propose optimal choice of the design parameters for random discontinuation designs (RDD) using a Bayesian decision-theoretic approach. We consider applications of RDDs to oncology phase II studies evaluating activity of cytostatic agents. The design consists of two stages. The preliminary open-label stage treats all patients with the new agent and identifies a possibly sensitive subpopulation. The subsequent second stage randomizes, treats, follows, and compares outcomes among patients in the identified subgroup, with randomization to either the new or a control treatment. Several tuning parameters characterize the design: the number of patients in the trial, the duration of the preliminary stage, and the duration of follow-up after randomization. We define a probability model for tumor growth, specify a suitable utility function, and develop a computational procedure for selecting the optimal tuning parameters.
Collapse
Affiliation(s)
- Lorenzo Trippa
- Harvard School of Public Health and Department of Biostatistics, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|
45
|
Clark PE. Rationale for targeted therapies and potential role of pazopanib in advanced renal cell carcinoma. Biologics 2010; 4:187-97. [PMID: 20714356 PMCID: PMC2921256 DOI: 10.2147/btt.s7818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2010] [Indexed: 11/23/2022]
Abstract
Advanced renal cell carcinoma (RCC) remains a challenging, major health problem. Recent advances in understanding the fundamental biology underlying one form of RCC, ie, clear cell (or conventional) RCC, have opened the door to a series of targeted agents, such as the tyrosine kinase inhibitors (TKIs), which have become the standard of care in managing advanced clear cell RCC. Among the newest of these agents to receive Food and Drug Administration approval in this disease is pazopanib. This review will summarize what is known about the fundamental biology that underlies clear cell RCC, the data surrounding the previously approved targeted agents for this disease, including not only the TKIs but also the mTOR inhibitors and the vascular endothelial growth factor-specific agent, bevacizumab, and the newest TKI, pazopanib. It will also explore the potential role for pazopanib relative to the other available agents and where it may fit into the armamentarium for treatment of advanced/metastatic RCC.
Collapse
Affiliation(s)
- Peter E Clark
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
46
|
Fernández A, Duch J, Flotats A, Camacho V, Estorch M, Carrió I. Evaluación de la respuesta terapéutica y PET-TAC: ¿realmente sólo importa el tamaño? ACTA ACUST UNITED AC 2010; 29:184-8. [DOI: 10.1016/j.remn.2010.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Accepted: 04/29/2010] [Indexed: 12/30/2022]
|
47
|
Ang MK, Tan SB, Lim WT. Phase II clinical trials in oncology: are we hitting the target? Expert Rev Anticancer Ther 2010; 10:427-38. [PMID: 20214523 DOI: 10.1586/era.09.178] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The number of novel and molecularly targeted agents in the last decade that need screening for preliminary efficacy in Phase II trials has increased. Many of these agents have a cytostatic mode of action that is difficult to assess using traditional Phase II designs. These new agents require detailed evaluation to optimize their dosing, to evaluate their effects on their target and to define early markers that predict for a definitive benefit. This review focuses on the options for Phase II trial designs. The different end points, single versus multiarm and randomized designs, the use of biomarkers and Bayesian approaches are also reviewed. The final design chosen will depend on the characteristics and circumstances of each individual study.
Collapse
Affiliation(s)
- Mei-Kim Ang
- National Cancer Centre Singapore, 11 Hospital Drive, Singapore.
| | | | | |
Collapse
|
48
|
Stewart DJ, Whitney SN, Kurzrock R. Equipoise Lost: Ethics, Costs, and the Regulation of Cancer Clinical Research. J Clin Oncol 2010; 28:2925-35. [PMID: 20406924 DOI: 10.1200/jco.2009.27.5404] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cancer is the leading cause of death in Americans younger than 85 years of age and kills one American every 56 seconds. Advances in understanding of cancer biology have given us the potential to develop new, effective targeted therapies. However, progress is slowed by suboptimal/outdated clinical trial design paradigms and by regulatory complexity and rigidity. For instance, simulations suggest that restricting randomized trials to patients expressing drug target, instead of using unselected patient populations, could substantially reduce patient numbers required to demonstrate efficacy. High response rates that are achievable when patients and drugs are matched on the basis of molecular profiles may also make some randomized trials unnecessary or unjustifiable. Moreover, increasing the regulatory rigidity of clinical trials (regulatory fundamentalism) augments trial complexity and costs while slowing progress without demonstrating meaningful safety benefits. Time from drug discovery to marketing increased from 8 years in 1960 to 12 to 15 years currently. Toxic death rates on phase I trials have decreased from 0.8% in 1979 to 0.5% by 2002, but the estimated cost per life-year gained by tighter regulations is $2,700,000 (far higher than costs of other health measures), and simulations suggest that regulatory delays in development of effective therapies result in tens to hundreds of thousands of life-years lost, whereas stringent regulations save extremely few. Dysregulation is also a major disincentive to patient and clinician participation in clinical research. In summary, current approaches squander research resources and discourage research participation, and the marked imbalance between potential life-years lost versus saved renders the regulatory burden potentially unethical. We outline suggested solutions.
Collapse
Affiliation(s)
- David J. Stewart
- From The University of Texas M. D. Anderson Cancer Center and Baylor College of Medicine, Houston, TX
| | - Simon N. Whitney
- From The University of Texas M. D. Anderson Cancer Center and Baylor College of Medicine, Houston, TX
| | - Razelle Kurzrock
- From The University of Texas M. D. Anderson Cancer Center and Baylor College of Medicine, Houston, TX
| |
Collapse
|
49
|
Abstract
A main criterion to identify activity in phase II studies is the response rates achieved in a well-defined subset of patients. The response could be defined as a measure of tumor shrinkage. For 30 years, metric methods have been used to assess this response. The World Health Organization was the first organization to propose a unified definition for response status. Over time, the latter evolved and 10 years ago an international consensus panel proposed the Response Evaluation Criteria in Solid Tumors criteria. Although these guidelines for response assessment have limitations and biases, they have nevertheless been proven useful and advantageous. This article reviews those criteria and describes their use.
Collapse
|
50
|
Abstract
Phase II oncologic clinical trials for agents that are mainly growth inhibitory and benefit only a selected patient population are challenging. The randomized discontinuation trial design is one approach by which this can be accomplished. A broad patient population is enrolled and all patients receive the investigational agent. Those with tumor shrinkage sufficient to be deemed of likely clinical significance after a prespecified period continue treatment, those with growth sufficient to be deemed clinically adverse and those with toxicity discontinue, and the remaining patients with "stable disease" are randomized to continue or discontinue therapy in a double-blind manner. The primary end point is the fraction of patients who remain progression free after an additional postrandomization period or the time to progression after randomization. By enriching for a possible sensitive population and then testing whether this was due to the agent or selection of an indolent disease group, the randomized discontinuation trial design efficiently assesses the putative growth inhibitory properties of an investigational agent and furthermore minimizes the number of patients exposed to placebo.
Collapse
|