1
|
Demirci RA, Gulati R, Hawley JE, Yezefski T, Haffner MC, Cheng HH, Montgomery RB, Schweizer MT, Yu EY, Nelson PS, Chen DL, Iravani A. SPECT/CT in Early Response Assessment of Patients with Metastatic Castration-Resistant Prostate Cancer Receiving 177Lu-PSMA-617. J Nucl Med 2024; 65:1945-1951. [PMID: 39510589 PMCID: PMC11937724 DOI: 10.2967/jnumed.124.267665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 10/15/2024] [Indexed: 11/15/2024] Open
Abstract
177Lu-PSMA-617 (LuPSMA) is a newly established treatment for patients with metastatic castration-resistant prostate cancer (mCRPC), but survival outcomes vary widely, and predictors of treatment responses are needed. This study investigated the role of total tumor volumes (TTVs) and new lesions (NLs) determined by LuPSMA SPECT/CT in early cycles to predict subsequent outcomes in a real-world practice setting. Methods: Between June and December 2022, consecutive patients with mCRPC who received at least 2 administrations of LuPSMA with SPECT/CT 24 h after treatment were retrospectively reviewed. We evaluated associations between TTVs and the appearance of NLs at cycles 2 and 3 with subsequent prostate-specific antigen (PSA) progression-free survival and overall survival (OS) using multivariate Cox regression. All analyses were adjusted for changes in PSA level relative to baseline. Results: Sixty-six mCRPC patients (median age, 74 y) received a median of 4 (interquartile range, 3-5) cycles of LuPSMA. Median follow-up starting at cycle 2 was 42 wk (interquartile range, 33-48 wk), with 24 of 66 patients deceased at the time of the analysis. Changes in TTV measured at the start of cycles 2 and 3 relative to baseline correlated significantly with corresponding changes in PSA level (r = 0.55 and 0.56), but absolute TTVs did not correlate significantly (r = 0.00 and 0.18). Patients with a higher absolute TTV at the start of cycle 2 had worse PSA progression-free survival and OS (hazard ratio [HR], 1.4 [95% CI, 1.1-1.8] and 2.1 [95% CI, 1.5-2.9]), with consistent results at the start of cycle 3 (HR, 2 [95% CI, 1.4-2.9] and 2 [95% CI, 1.2-3.2]). NLs were identified in 13 of 66 and 11 of 51 patients at the start of cycles 2 and 3. NLs at the start of cycle 2 were associated with worse OS (HR, 5.8 [95% CI, 1.9-17.5]), with consistent results at the start of cycle 3 (HR, 4.9 [95% CI, 1.3-18.6]). In multivariate analysis, a higher TTV and the appearance of NLs at the start of cycles 2 and 3 were independently associated with poorer OS. Conclusion: Higher TTVs and NLs on LuPSMA SPECT/CT at the start of cycles 2 and 3 were independently associated with higher risk of death. These measures provided prognostic information independent of changes in PSA. Development of prognostic and predictive models including TTV, NLs, and PSA changes is warranted.
Collapse
Affiliation(s)
| | - Roman Gulati
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Jessica E Hawley
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, Washington
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Todd Yezefski
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, Washington
| | - Michael C Haffner
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington; and
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Heather H Cheng
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, Washington
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Robert B Montgomery
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, Washington
| | - Michael T Schweizer
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, Washington
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Evan Y Yu
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, Washington
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Peter S Nelson
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, Washington
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington; and
| | - Delphine L Chen
- Department of Radiology, University of Washington, Seattle, Washington
| | - Amir Iravani
- Department of Radiology, University of Washington, Seattle, Washington;
| |
Collapse
|
2
|
Xia Y, Zhou J, Xun X, Johnston L, Wei T, Gao R, Zhang Y, Reddy B, Liu C, Kim G, Zhang J, Zhao S, Yu Z. Deep learning for oncologic treatment outcomes and endpoints evaluation from CT scans in liver cancer. NPJ Precis Oncol 2024; 8:263. [PMID: 39551847 PMCID: PMC11570623 DOI: 10.1038/s41698-024-00754-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 11/04/2024] [Indexed: 11/19/2024] Open
Abstract
Accurate treatment response assessment using serial CT scans is essential in oncological clinical trials. However, oncologists' assessment following the Response Evaluation Criteria in Solid Tumors (RECIST) guideline is subjective, time-consuming, and sometimes fallible. Advanced liver cancer often presents multifocal hepatic lesions on CT imaging, making accurate characterization more challenging than with other malignancies. In this work, we developed a tumor volume guided comprehensive objective response evaluation based on deep learning (RECORD) for liver cancer. RECORD performs liver tumor segmentation, followed by sum of the volume (SOV)-based treatment response classification and new lesion assessment. Then, it can provide treatment evaluations of response, stability, and progression, and calculates progression-free survival (PFS) and response time. The RECORD pipeline was developed with both CNN and ViT backbones. Its performance was evaluated in three longitudinal cohorts involving 60 multi-national centers, 206 patients, 891 CT scans, using internal five-fold cross-validation and external validations. RECORD with the most effective backbone achieved an average AUC-response of 0.981, AUC-stable of 0.929, and AUC-progression of 0.969 for SOV-based disease status classification, F1-score of 0.887 for new lesion identification, and accuracy of 0.889 for final treatment outcome assessments across all cohorts. RECORD's PFS and response time predictions strongly correlated with clinician's assessments (P < 0.001). Moreover, RECORD can better stratify high-risk versus low-risk patients for overall survival compared to the human-assessed RECIST results. In conclusion, RECORD demonstrates efficiency and objectivity in analyzing liver lesions for treatment response evaluation. Further research should extend the pipeline to other metastatic organ sites.
Collapse
Affiliation(s)
- Yujia Xia
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
- SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jie Zhou
- SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University, Shanghai, 200240, China
- Department of Statistics, School of Mathematical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaolei Xun
- Statistics in Global Statistics and Data Science, Beigene, Shanghai, 200040, China
| | - Luke Johnston
- SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University, Shanghai, 200240, China
- Department of Statistics, School of Mathematical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ting Wei
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
- SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ruitian Gao
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
- SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yufei Zhang
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
- SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Bobby Reddy
- PiHealth USA, 55 Cambridge Parkway, Cambridge, MA, 02142, USA
| | - Chao Liu
- PiHealth USA, 55 Cambridge Parkway, Cambridge, MA, 02142, USA
| | - Geoffrey Kim
- PiHealth USA, 55 Cambridge Parkway, Cambridge, MA, 02142, USA
| | - Jin Zhang
- PiHealth USA, 55 Cambridge Parkway, Cambridge, MA, 02142, USA.
| | - Shuai Zhao
- Department of Transplantation, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Zhangsheng Yu
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China.
- SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University, Shanghai, 200240, China.
- Department of Statistics, School of Mathematical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China.
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
3
|
Yadav S, Lowery B, Tuchayi AM, Jiang F, Saelee R, Aggarwal RR, Juarez R, Flavell RR, Hope TA. Impact of Posttreatment SPECT/CT on Patient Management During 177Lu-PSMA-617 Radiopharmaceutical Therapy. J Nucl Med 2024; 65:1395-1401. [PMID: 39117452 DOI: 10.2967/jnumed.124.267955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/04/2024] [Indexed: 08/10/2024] Open
Abstract
177Lu can be imaged after administration using SPECT/CT. Most work to date has focused on using posttreatment imaging to measure normal organ and tumor dose. We aimed to assess the impact of posttreatment SPECT/CT on the management of patients undergoing 177Lu-prostate-specific membrane antigen (PSMA) radiopharmaceutical therapy (RPT). Methods: In this retrospective study, 122 patients underwent PSMA RPT with subsequent SPECT/CT 24 h after treatment. We determined a qualitative response at each cycle and reviewed patient charts to assess the impact that posttreatment SPECT/CT had on patient management. Changes in patient management were classified as changes on the basis of progression and response, and specific cycles when they occurred were noted. Miscellaneous changes in patient management were also evaluated. Results: Among the 122 consecutive patients examined, 42%-56% exhibited stable disease, whereas 19%-39% of patients exhibited response on visual assessment across treatment cycles. In total, 49% (n = 60) of patients experienced changes in management, of which 57% (n = 34) were due to progression, 40% (n = 24) were due to response, and 3% (n = 2) were due to miscellaneous changes. Changes due to disease progression were observed mostly after cycles 2 and 4. Changes due to response to RPT occurred mostly after cycles 3 and 4. Conclusion: At our center, 49% of patients experienced changes in management based on posttreatment SPECT/CT, and most of these changes occurred at cycles 2 and 4. Integrating posttreatment SPECT/CT into routine PSMA RPT protocols can aid in patient management.
Collapse
Affiliation(s)
- Surekha Yadav
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Blair Lowery
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Abuzar Moradi Tuchayi
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Fei Jiang
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California
| | - Rachelle Saelee
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Rahul R Aggarwal
- Division of Medical Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Roxanna Juarez
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California; and
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California;
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California; and
- Department of Radiology, San Francisco VA Medical Center, San Francisco, California
| |
Collapse
|
4
|
Li R, Zhou Q, Liu G, Zhu F, Liu Z, Bo H, Fan L. CSNK1G2-AS1 promotes metastasis, colony formation and serves as a biomarker in testicular germ cell tumor cells. J Cancer 2023; 14:2771-2783. [PMID: 37781070 PMCID: PMC10539554 DOI: 10.7150/jca.85640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/09/2023] [Indexed: 10/03/2023] Open
Abstract
Background/Aim: Some long non-coding RNAs (lncRNAs) have been found to significantly participate in the progression of TGCTs. In comparison to the normal testis, the TGCT tissues showed significantly decreased CSNK1G2-AS1 expression, however, its effect on TGCTs and its mechanism are still unclear. The aim of this study is to investigate the effect of CSNK1G2-AS1 on TGCTs and explore the mechanism underlying its effect on TGCTs. Materials and Methods: In this study, to evaluate the expression of CSNK1G2-AS1 in tissue samples from TGCTs, the UCSC and GEPIA databases were applied and qRT-PCR was conducted. The Kaplan-Meier Plotter was applied to analyze the correlation between CSNK1G2-AS1 methylation levels and the prognosis of TGCTs patients. The assays of MTS, clone formation, transwell, and flow cytometry were performed to investigate the effect of CSNK1G2-AS1 overexpression on the proliferation, metastasis, and apoptosis of TGCT cells, respectively. Finally, western blotting was conducted to determine the expressions of the proteins associated with EMT and AKT. Results: Our study first found that, compared to the normal testis, TGCTs tissue showed significantly decreased CSNK1G2-AS1 expression, and hypomethylation of CSNK1G2-AS1 was significantly correlated with a better prognosis with TGCTs patients. In vitro, we found that overexpression of CSNK1G2-AS1 dramatically promoted the clone formation, invasion, and migration of TGCT cells, but inhibited apoptosis. And CSNK1G2-AS1 overexpression significantly decreased the expression of EMT-associated proteins ZO-1 but increased the expression and phosphorylation of AKT. Conclusions: CSNK1G2-AS1 may play an essential role in the pathogenesis and metastasis of TGCTs through the EMT- and AKT-mediated signal pathways.
Collapse
Affiliation(s)
- Ruixue Li
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410078, People's Republic of China
| | - Qianyin Zhou
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410078, People's Republic of China
| | - Guangmin Liu
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410078, People's Republic of China
| | - Fang Zhu
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410078, People's Republic of China
| | - Zhizhong Liu
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410078, People's Republic of China
- Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410078, People's Republic of China
| | - Hao Bo
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410078, People's Republic of China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, People's Republic of China
| | - Liqing Fan
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410078, People's Republic of China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, People's Republic of China
| |
Collapse
|
5
|
John N, Pathmanandavel S, Crumbaker M, Counter W, Ho B, Yam AO, Wilson P, Niman R, Ayers M, Poole A, Hickey A, Agrawal S, Perkins G, Kallinen A, Eslick E, Stockler MR, Joshua AM, Nguyen A, Emmett L. 177Lu-PSMA SPECT Quantitation at 6 Weeks (Dose 2) Predicts Short Progression-Free Survival for Patients Undergoing 177Lu-PSMA-I&T Therapy. J Nucl Med 2023; 64:410-415. [PMID: 36215568 DOI: 10.2967/jnumed.122.264677] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/01/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
177Lu-PSMA is an effective treatment in metastatic castration-resistant prostate cancer (mCRPC). Our ability to assess response rates and adjust treatment may be improved using predictive tools. This study aimed to evaluate change in 177Lu-PSMA SPECT quantitative parameters to monitor treatment response. Methods: One hundred twenty-seven men with progressive mCRPC previously treated with androgen-signaling inhibition (99%) and chemotherapy (71%) received a median of 3 (interquartile range [IQR], 2-5) 8-GBq (IQR, 8-8.5 GBq) doses of 177Lu-PSMA-I&T. Imaging included 68Ga-PSMA-11 PET/CT (SUVmax > 15 at a single site and > 10 at all sites > 2 cm), diagnostic CT, and 177Lu SPECT/CT from vertex to mid thigh (24 h after treatment). 177Lu SPECT/CT quantitative analysis was undertaken at cycles 1 (baseline) and 2 (week 6) of treatment. Clinical and biochemical results were assessed to evaluate prostate-specific antigen (PSA) progression-free survival (PFS) and overall survival (OS). Results: A PSA reduction of more than 50% was seen in 58% (74/127). The median PSA PFS was 6.1 mo (95% CI, 5.5-6.7), and OS was 16.8 mo (95% CI, 13.5-20.1). At the time of analysis, 41% (52/127) were deceased. At baseline and week 6, 76% (96/127) had analyzable serial 177Lu SPECT/CT imaging. SPECT total tumor volume (TTV) was reduced between baseline and week 6 in 74% (71/96; median, -193; IQR, -486 to -41). Any increase in SPECT TTV between baseline and week 6 was associated with significantly shorter PSA PFS (hazard ratio, 2.5; 95% CI, 1.5-4.2; P = 0.0008) but not OS. Median PSA PFS in those with an increase in SPECT TTV was 3.7 mo (95% CI, 2.8-6.8), compared with 6.7 mo (95% CI, 5.8-10.6) in those with no increase in SPECT TTV. An increase in SPECT TTV greater than 20% was also associated with PSA PFS (hazard ratio, 1.9; 95% CI, 1.2-3.0; P = 0.008) but less significantly than any change in SPECT TTV. There was a significant difference in PSA PFS between patients with both increased PSA and SPECT TTV and patients with reduced SPECT TTV and PSA (median, 2.8 vs. 9.0 mo; P < 0.0001). Conclusion: Increasing PSMA SPECT TTV on quantitative 177Lu SPECT/CT predicts short progression-free survival and may play a future role as an imaging response biomarker, identifying when to cease or intensify 177Lu-PSMA therapy.
Collapse
Affiliation(s)
- Nikeith John
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Sarennya Pathmanandavel
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | - Megan Crumbaker
- St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Kinghorn Cancer Centre, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | - William Counter
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | - Bao Ho
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | - Andrew O Yam
- St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Kinghorn Cancer Centre, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | | | - Remy Niman
- MIM Software, Inc., Cleveland, Ohio; and
| | - Maria Ayers
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | - Aron Poole
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | - Adam Hickey
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | - Shikha Agrawal
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | - Gary Perkins
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | - Annukka Kallinen
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | - Enid Eslick
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | - Martin R Stockler
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Anthony M Joshua
- St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Kinghorn Cancer Centre, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | - Andrew Nguyen
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, New South Wales, Australia;
- St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| |
Collapse
|
6
|
Pathmanandavel S, Crumbaker M, Ho B, Yam AO, Wilson P, Niman R, Ayers M, Sharma S, Hickey A, Eu P, Stockler M, Martin AJ, Joshua AM, Nguyen A, Emmett L. Evaluation of 177Lu-PSMA-617 SPECT/CT Quantitation as a Response Biomarker Within a Prospective 177Lu-PSMA-617 and NOX66 Combination Trial (LuPIN). J Nucl Med 2023; 64:221-226. [PMID: 36008120 PMCID: PMC9902857 DOI: 10.2967/jnumed.122.264398] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 02/04/2023] Open
Abstract
177Lu-PSMA-617 is an effective and novel treatment in metastatic castration-resistant prostate cancer (mCRPC). Our ability to assess response rates and therefore efficacy may be improved using predictive tools. This study investigated the predictive value of serial 177Lu-PSMA-617 SPECT/CT (177Lu SPECT) imaging in monitoring treatment response. Methods: Fifty-six men with progressive mCRPC previously treated with chemotherapy and novel androgen signaling inhibitor were enrolled into the LuPIN trial and received up to 6 doses of 177Lu-PSMA-617 and a radiation sensitizer (3-(4-hydroxyphenyl)-2H-1-benzopyran-7-ol [NOX66]). 68Ga-PSMA-11 and 18F-FDG PET/CT were performed at study entry and exit, and 177Lu SPECT from vertex to mid thighs was performed 24 h after each treatment. SPECT quantitative analysis was undertaken at cycles 1 (baseline) and 3 (week 12) of treatment. Results: Thirty-two of the 56 men had analyzable serial 177Lu SPECT imaging at both cycle 1 and cycle 3. In this subgroup, median prostate-specific antigen (PSA) progression-free survival (PFS) was 6.3 mo (95% CI, 5-10 mo) and median overall survival was 12.3 mo (95% CI, 12-24 mo). The PSA 50% response rate was 63% (20/32). 177Lu SPECT total tumor volume (SPECT TTV) was reduced in 68% (22/32; median, -0.20 m3 [95% CI, -1.4 to -0.001]) and increased in 31% (10/32; median, 0.36 [95% CI, 0.1-1.4]). Any increase in SPECT TTV was associated with shorter PSA PFS (hazard ratio, 4.1 [95% CI, 1.5-11.2]; P = 0.006). An increase of 30% or more in SPECT TTV was also associated with a shorter PSA PFS (hazard ratio, 3.3 [95% CI, 1.3-8.6]; P =0.02). Tumoral SUVmax was reduced in 91% (29/32) and SUVmean in 84% (27/32); neither was associated with PSA PFS or overall survival outcomes. PSA progression by week 12 was also associated with a shorter PSA PFS (hazard ratio, 26.5 [95% CI, 5.4-131]). In the patients with SPECT TTV progression at week 12, 50% (5/10) had no concurrent PSA progression (median PSA PFS, 4.5 mo [95% CI, 2.8-5.6 mo]), and 5 of 10 men had both PSA and SPECT TTV progression at week 12 (median PSA PFS, 2.8 mo [95% CI, 1.8-3.7 mo]). Conclusion: Increasing SPECT TTV on quantitative 177Lu SPECT predicts a short PFS and may play a future role as an imaging response biomarker.
Collapse
Affiliation(s)
- Sarennya Pathmanandavel
- Department of Theranostics and Nuclear Medicine, St. Vincent’s Hospital, Sydney, New South Wales, Australia
| | - Megan Crumbaker
- Kinghorn Cancer Centre, St. Vincent’s Hospital, Sydney, New South Wales, Australia;,Garvan Institute of Medical Research, Sydney, New South Wales, Australia;,St. Vincent’s Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Bao Ho
- Department of Theranostics and Nuclear Medicine, St. Vincent’s Hospital, Sydney, New South Wales, Australia
| | - Andrew O. Yam
- Kinghorn Cancer Centre, St. Vincent’s Hospital, Sydney, New South Wales, Australia;,Garvan Institute of Medical Research, Sydney, New South Wales, Australia;,St. Vincent’s Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | | | | | - Maria Ayers
- Department of Theranostics and Nuclear Medicine, St. Vincent’s Hospital, Sydney, New South Wales, Australia
| | - Shikha Sharma
- Department of Theranostics and Nuclear Medicine, St. Vincent’s Hospital, Sydney, New South Wales, Australia
| | - Adam Hickey
- Department of Theranostics and Nuclear Medicine, St. Vincent’s Hospital, Sydney, New South Wales, Australia
| | - Peter Eu
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and
| | - Martin Stockler
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Andrew J. Martin
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Anthony M. Joshua
- Kinghorn Cancer Centre, St. Vincent’s Hospital, Sydney, New South Wales, Australia;,Garvan Institute of Medical Research, Sydney, New South Wales, Australia;,St. Vincent’s Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Andrew Nguyen
- Department of Theranostics and Nuclear Medicine, St. Vincent’s Hospital, Sydney, New South Wales, Australia;,St. Vincent’s Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, New South Wales, Australia; .,Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
7
|
Ching JCF, Lam S, Lam CCH, Lui AOY, Kwong JCK, Lo AYH, Chan JWH, Cai J, Leung WS, Lee SWY. Integrating CT-based radiomic model with clinical features improves long-term prognostication in high-risk prostate cancer. Front Oncol 2023; 13:1060687. [PMID: 37205204 PMCID: PMC10186349 DOI: 10.3389/fonc.2023.1060687] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/10/2023] [Indexed: 05/21/2023] Open
Abstract
Objective High-risk prostate cancer (PCa) is often treated by prostate-only radiotherapy (PORT) owing to its favourable toxicity profile compared to whole-pelvic radiotherapy. Unfortunately, more than 50% patients still developed disease progression following PORT. Conventional clinical factors may be unable to identify at-risk subgroups in the era of precision medicine. In this study, we aimed to investigate the prognostic value of pre-treatment planning computed tomography (pCT)-based radiomic features and clinical attributes to predict 5-year progression-free survival (PFS) in high-risk PCa patients following PORT. Materials and methods A total of 176 biopsy-confirmed PCa patients who were treated at the Hong Kong Princess Margaret Hospital were retrospectively screened for eligibility. Clinical data and pCT of one hundred eligible high-risk PCa patients were analysed. Radiomic features were extracted from the gross-tumour-volume (GTV) with and without applying Laplacian-of-Gaussian (LoG) filter. The entire patient cohort was temporally stratified into a training and an independent validation cohort in a ratio of 3:1. Radiomics (R), clinical (C) and radiomic-clinical (RC) combined models were developed by Ridge regression through 5-fold cross-validation with 100 iterations on the training cohort. A model score was calculated for each model based on the included features. Model classification performance on 5-year PFS was evaluated in the independent validation cohort by average area-under-curve (AUC) of receiver-operating-characteristics (ROC) curve and precision-recall curve (PRC). Delong's test was used for model comparison. Results The RC combined model which contains 6 predictive features (tumour flatness, root-mean-square on fine LoG-filtered image, prostate-specific antigen serum concentration, Gleason score, Roach score and GTV volume) was the best-performing model (AUC = 0.797, 95%CI = 0.768-0.826), which significantly outperformed the R-model (AUC = 0.795, 95%CI = 0.774-0.816) and C-model (AUC = 0.625, 95%CI = 0.585-0.665) in the independent validation cohort. Besides, only the RC model score significantly classified patients in both cohorts into progression and progression-free groups regarding their 5-year PFS (p< 0.05). Conclusion Combining pCT-based radiomic and clinical attributes provided superior prognostication value regarding 5-year PFS in high-risk PCa patients following PORT. A large multi-centre study will potentially aid clinicians in implementing personalised treatment for this vulnerable subgroup in the future.
Collapse
Affiliation(s)
- Jerry C. F. Ching
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Saikit Lam
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Research Institute for Smart Aging, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Cody C. H. Lam
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Angie O. Y. Lui
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Joanne C. K. Kwong
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Anson Y. H. Lo
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Jason W. H. Chan
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Jing Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - W. S. Leung
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- *Correspondence: Shara W. Y. Lee, ; W. S. Leung,
| | - Shara W. Y. Lee
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- *Correspondence: Shara W. Y. Lee, ; W. S. Leung,
| |
Collapse
|
8
|
Joshua AM, Armstrong A, Crumbaker M, Scher HI, de Bono J, Tombal B, Hussain M, Sternberg CN, Gillessen S, Carles J, Fizazi K, Lin P, Duggan W, Sugg J, Russell D, Beer TM. Statin and metformin use and outcomes in patients with castration-resistant prostate cancer treated with enzalutamide: A meta-analysis of AFFIRM, PREVAIL and PROSPER. Eur J Cancer 2022; 170:285-295. [PMID: 35643841 PMCID: PMC10394474 DOI: 10.1016/j.ejca.2022.04.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Statins and metformin are commonly prescribed for patients, including those with prostate cancer. Preclinical and epidemiologic studies of each agent have suggested anti-cancer properties. METHODS Patient data from three randomised, double-blind, placebo-controlled, phase III studies evaluating enzalutamide (AFFIRM, PREVAIL and PROSPER) in patients with castration-resistant prostate cancer were included in this analysis. This post hoc, retrospective study examined the association of statin and metformin on radiographic progression-free survival (rPFS), metastasis-free survival (MFS), toxicity and overall survival (OS). After adjusting for available clinical prognostic variables, multivariate analyses were performed on pooled data from AFFIRM and PREVAIL, all three trials pooled, and each trial individually, to assess differential efficacy in these end-points associated with the baseline use of these medications. RESULTS In the multivariate analysis of the individual trials, OS and rPFS/MFS were not significantly influenced by statin or metformin use in AFFIRM or PROSPER. However, in PREVAIL, OS was significantly influenced by statin (hazard ratio [HR] 0.72; 95% confidence interval [CI] 0.59-0.89) and rPFS was significantly influenced by metformin (HR, 0.48; 95% CI 0.34-0.70). In pooled analyses, improved OS was significantly associated with statin use but not metformin use for AFFIRM+PREVAIL trials (HR 0.83; 95% CI 0.72-0.96) and AFFIRM+PREVAIL+PROSPER (HR 0.75; 95% CI 0.66-0.85). CONCLUSIONS The association between statin or metformin use and rPFS, MFS and OS was inconsistent across three trials. Analyses of all three trials pooled and AFFIRM+PREVAIL pooled revealed that statin but not metformin use was significantly associated with a reduced risk of death in enzalutamide-treated patients. Additional prospective, controlled studies are warranted. CLINICAL TRIAL REGISTRATION AFFIRM (NCT00974311), PREVAIL (NCT01212991) and PROSPER (NCT02003924).
Collapse
Affiliation(s)
- Anthony M Joshua
- Kinghorn Cancer Centre, St. Vincent's Hospital, Sydney, NSW, Australia.
| | - Andrew Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC, USA
| | - Megan Crumbaker
- Kinghorn Cancer Centre, St. Vincent's Hospital, Sydney, NSW, Australia
| | - Howard I Scher
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Johann de Bono
- The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | | | - Maha Hussain
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Cora N Sternberg
- Englander Institute for Precision Medicine, Weill Cornell Medicine, Meyer Cancer Center, New York, NY, USA
| | - Silke Gillessen
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
| | - Joan Carles
- Vall D'Hebron University Hospital, Vall D'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Karim Fizazi
- Institut Gustave Roussy, University of Paris Saclay, Villejuif, France
| | - Ping Lin
- Formerly of Pfizer Inc., San Francisco, CA, USA
| | | | | | | | - Tomasz M Beer
- OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
9
|
Qin Z, Liu K, Xu X, Li T, Ge Y, Wu B, Xing C, Mao H. Incidence, predictors and 6-month overall outcome of acute kidney injury in Chinese patients receiving PD-1 inhibitors. Future Oncol 2022; 18:1951-1962. [PMID: 35232231 DOI: 10.2217/fon-2021-1004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Aim: To explore the incidence, risk factors and overall outcome of the first episode of immune checkpoint inhibitor-related acute kidney injury (ICI-AKI) in Chinese patients receiving PD-1 inhibitors. Methods: Data for patients receiving PD-1 inhibitors at Jiangsu Province Hospital between December 2017 and January 2020 were retrospectively reviewed. Results: A total of 5.6% of 551 patients receiving PD-1 inhibitors developed ICI-AKI. Concomitant use of nonsteroidal anti-inflammatory drugs, ICI cycles and extrarenal immune-related adverse events may be independently associated with ICI-AKI. ICI-AKI may not be a risk factor for increased mortality or worse progression-free survival. Conclusions: ICI-AKI is relatively rare and its occurrence may not affect the overall six-month outcome of patients receiving PD-1 inhibitors. Further studies are needed to verify these findings.
Collapse
Affiliation(s)
- Zhongke Qin
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing, 210029, China
| | - Kang Liu
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing, 210029, China
| | - Xueqiang Xu
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing, 210029, China
| | - Ting Li
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing, 210029, China
| | - Yifei Ge
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing, 210029, China
| | - Buyun Wu
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing, 210029, China
| | - Changying Xing
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing, 210029, China
| | - Huijuan Mao
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing, 210029, China
| |
Collapse
|
10
|
hsa_circ_0001275 Is One of a Number of circRNAs Dysregulated in Enzalutamide Resistant Prostate Cancer and Confers Enzalutamide Resistance In Vitro. Cancers (Basel) 2021; 13:cancers13246383. [PMID: 34945002 PMCID: PMC8715667 DOI: 10.3390/cancers13246383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/07/2021] [Indexed: 01/22/2023] Open
Abstract
Simple Summary Although newer generations of androgen deprivation therapy such as enzalutamide are providing hope, it is clinically challenging to deliver effective therapy to individuals with metastatic castrate-resistant prostate cancer. Between 20–40% of patients have intrinsic resistance to therapy and all patients will ultimately experience disease progression due to acquired resistance, which is a significant clinical dilemma. The aim of our study was to evaluate the role of circular RNAs (circRNAs) in enzalutamide-resistant prostate cancer as part of the effort to identify useful biomarkers for patient selection and potential new therapeutic targets. We confirmed that hsa_circ_0001275 was highly upregulated in an enzalutamide resistant cell line and demonstrated that its overexpression resulted in increased enzalutamide resistance. Our data showed that hsa_circ_0001275 was not expressed abundantly in patient plasma samples, however, a trend of expression was evident which paralleled disease activity indicating a possible association with enzalutamide resistance. Overall, we have provided evidence that hsa_circ_0001275 promotes enzalutamide resistance and thus may serve as a potential therapeutic target. Abstract Background: Enzalutamide is part of the treatment regimen for metastatic castration-resistant prostate cancer (MCRPC). However, both intrinsic and acquired resistance to the drug remain substantial clinical quandaries. circRNAs, a novel type of non-coding RNA, have been identified in a number of cancers including prostate cancer and have been associated with cancer development and progression. circRNAs have shown great potential as clinically useful blood-based ‘liquid biopsies’ and as therapeutic targets in prostate cancer. The aim of this study was to examine the role of circRNA transcripts in enzalutamide-resistant prostate cancer cells and assess their utility as biomarkers. Methods: An isogenic cell line model of enzalutamide resistance was subjected to circRNA microarray profiling. Several differentially expressed circRNAs, along with their putative parental genes were validated using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). circRNAs of interest were stably overexpressed in the control cell line and drug sensitivity was assessed using an ELISA-based proliferation assay. The candidate circRNA, hsa_circ_0001275, was measured in patient plasma samples using RT-droplet digital PCR (RT-ddPCR). Results: hsa_circ_0001275 and its parental gene, PLCL2, were significantly up-regulated in strongly resistant clones vs. control (p < 0.05). Overexpression of hsa_circ_0001275 in the control cell line resulted in increased resistance to enzalutamide (p < 0.05). While RT-ddPCR analysis of hsa_circ_0001275 expression in plasma samples of 44 clinical trial participants showed a trend that mirrored the stages of disease activity (as defined by PSA level), the association did not reach statistical significance. Conclusions: Our data suggest that increased levels of hsa_circ_0001275 contribute to enzalutamide resistance. hsa_circ_0001275 plasma expression showed a trend that mirrors the PSA level at specific disease time points, indicating that circRNAs mirror disease recurrence and burden and may be associated with enzalutamide resistance.
Collapse
|
11
|
Maj-Hes A, Szarvas T, Sevcenco S, Kramer G. Multiple Docetaxel Retreatments Without Prednisone for Metastatic Castration-Resistant Prostate Cancer in the Docetaxel-Only Era: Effects on PSA Kinetics and Survival. Adv Ther 2021; 38:3831-3841. [PMID: 34043207 PMCID: PMC8280015 DOI: 10.1007/s12325-021-01778-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 05/06/2021] [Indexed: 01/10/2023]
Abstract
Introduction This study aimed to assess the effects of multiple docetaxel (DOC) treatments on prostate-specific antigen (PSA) kinetics and survival among patients with metastatic castration-resistant prostate cancer (mCRPC) who were sensitive to first-line DOC and received no other life-prolonging agents. To eliminate the effect of cortisone on serum PSA, only patients who were treated without prednisone were included. Methods This IRB-approved retrospective study evaluated 52 patients with mCRPC who were retreated using DOC after first-line DOC (without prednisone in both cases), based on a PSA response of > 50% and no radiographic progression. Twenty-three PSA-based factors, including static and kinetic PSA measures, were evaluate for their ability to predict overall survival (OS) Results The patients received 688 cycles of DOC in 143 series, including 91 courses of retreatments (1 cycle: 28 patients, 2 cycles: 14 patients, 3 cycles: 8 patients, 4 cycles: 1 patient, and 7 cycles: 1 patient). The median overall number of cycles per patient was 12 (range: 7–31). The median durations of the first, second, and third holidays were 18 weeks (6–60 weeks), 16 weeks (3–44 weeks), and 17 weeks (8–51 weeks), respectively. The median OSs were 22 months (10.5–70 months) after the first DOC treatment and 14 months (3–65 months) after the second DOC treatment. The > 50% PSA decline rate was 48% after retreatment. Short treatment holidays (< 3 months) were associated with shortened OS (p = 0.01). In the multivariate analysis, a 25% PSA increase over the nadir was the strongest predictor of survival (HR: 3.20, 95% CI: 1.47–6.99, p = 0.003). Conclusions DOC retreatment without prednisone had anti-tumor activity in a considerable proportion of mCRPC cases that were initially sensitive to first-line DOC. A 25% PSA increase over the nadir might predict acquired DOC resistance.
Collapse
|
12
|
Xie S, Sun W, Zhang C, Dong B, Yang J, Hou M, Xiong L, Cai B, Liu X, Xue W. Metabolic Control by Heat Stress Determining Cell Fate to Ferroptosis for Effective Cancer Therapy. ACS NANO 2021; 15:7179-7194. [PMID: 33861924 DOI: 10.1021/acsnano.1c00380] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Flexible manipulation of the fate of cancer cells through exogenous stimulation-induced metabolic reprogramming could handle the cellular plasticity-derived therapies resistance, which provides an effective paradigm for the treatment of refractory and relapsing tumors in clinical settings. Herein, we demonstrated that moderate heat (45 °C) could significantly regress the expression of antioxidants and trigger specific lipid metabolic reprogramming in cancer cells synergized with iron oxide nanoparticles (Fe3O4 NPs). This metabolic control behavior destroyed the tumor redox homeostasis and produced overwhelming lipid peroxides, consequently sensitizing the tumor to ferroptosis. Based on these findings, a heat-triggered tumor-specific ferroptosis strategy was proposed by the rational design of a polypeptide-modified and 1H-perfluoropentane (1H-PFP)-encapsulated Fe3O4-containing nanoformulation (GBP@Fe3O4). When irradiated by an 808 nm laser, the phase transition of 1H-PFP was triggered by localized moderate heat (45 °C), leading to burst release of Fe3O4in situ to produce potent reactive oxygen species through the Fenton reaction in the tumor microenvironment. Together with the antioxidant inhibition response and distinctive lipid metabolic reprogramming by heat stress, this oxidative damage was amplified to induce tumor ferroptosis and achieve sufficient antitumor effects. Importantly, we confirmed that ACSBG1, an acyl-CoA synthetase, was the key pro-ferroptotic factor in this heat-induced ferroptosis process. Moreover, knockout of this gene could realize cancer cell death fate conversion from ferroptosis to non-ferroptotic death. This work provides mechanistic insights and practical strategies for heat-triggered ferroptosis in situ to reduce the potential side effects of direct ferroptosis inducers and highlights the key factor in regulating cell fate under heat stress.
Collapse
Affiliation(s)
- Shaowei Xie
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Department of Ultrasound, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wenshe Sun
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Chunfu Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Baijun Dong
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jingxing Yang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Mengfei Hou
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Liqin Xiong
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Biao Cai
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xuesong Liu
- Department of Ultrasound, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wei Xue
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
13
|
Prostate-specific Antigen Progression in Enzalutamide-treated Men with Nonmetastatic Castration-resistant Prostate Cancer: Any Rise in Prostate-specific Antigen May Require Closer Monitoring. Eur Urol 2020; 78:847-853. [DOI: 10.1016/j.eururo.2020.08.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 08/16/2020] [Indexed: 11/23/2022]
|
14
|
Abufaraj M, Iwata T, Kimura S, Haddad A, Al-Ani H, Abusubaih L, Moschini M, Briganti A, Karakiewicz PI, Shariat SF. Differential Impact of Gonadotropin-releasing Hormone Antagonist Versus Agonist on Clinical Safety and Oncologic Outcomes on Patients with Metastatic Prostate Cancer: A Meta-analysis of Randomized Controlled Trials. Eur Urol 2020; 79:44-53. [PMID: 32605859 DOI: 10.1016/j.eururo.2020.06.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/02/2020] [Indexed: 12/24/2022]
Abstract
CONTEXT Androgen deprivation therapy is the mainstay treatment of metastatic prostate cancer, achieved mainly by gonadotropin-releasing hormone (GnRH) agonists or antagonists. OBJECTIVE To investigate the differential impact of GnRH agonists and antagonists on clinical safety and oncologic outcomes. EVIDENCE ACQUISITION This meta-analysis was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines. A literature search using the electronic databases (MEDLINE, Web of Science, Cochrane Library, and Scopus) included randomized controlled trials comparing the clinical safety and oncologic outcomes of GnRH agonists and antagonists. The endpoints of interest were the following: (1) treatment-related adverse effects (AEs), (2) prostate-specific antigen (PSA) progression, and (3) overall mortality. The relative risk (RR) was used as the summary statistic, and results were reported with 95% confidence intervals (CIs). EVIDENCE SYNTHESIS Eight clinical trials (20 published studies) comprising 2632 men met our inclusion criteria; of them, 1646 received GnRH antagonist and 986 had GnRH agonist. Treatment-emerging AEs occurred in 73% patients in the GnRH antagonist group and 68% in the GnRH agonist group (RR: 1.10, 95% CI: 1.04-1.15). Serious AEs occurred in 9.8% of the GnRH antagonist and 11% of the GnRH agonist group (RR: 0.92, 95% CI: 0.73-1.17). Antagonists were associated with higher injection site reaction rates (38%) than agonists (4.8%). GnRH antagonist was associated with fewer cardiovascular events (RR: 0.52, 95% CI: 0.34-0.80). There was no significant difference in PSA progression, but GnRH antagonist was associated with lower overall mortality rates than GnRH agonists (RR: 0.48, 95% CI: 0.26-0.90, p = 0.02). CONCLUSIONS Existing data indicate that GnRH antagonist use is associated with significantly lower overall mortality and cardiovascular events as compared with agonists. These findings should be interpreted with caution owing to the short follow-up duration and assessment of cardiovascular events as secondary endpoints in the included trials. Further studies are needed to validate or refute these observations. Injection site reactions were significantly higher in the GnRH antagonist group. PATIENT SUMMARY Gonadotropin-releasing hormone (GnRH) antagonist is associated with lower death rates and cardiovascular events than GnRH agonists, based on the data from trials with short follow-up duration. GnRH agonists are associated with lower adverse events, such as decreased libido, hot flushes, erectile dysfunction, back pain, weight gain, constipation, and injection site reactions. There were no significant differences in prostate-specific antigen progression or fatigue.
Collapse
Affiliation(s)
- Mohammad Abufaraj
- Department of Special Surgery, Jordan University Hospital, The University of Jordan, Amman, Jordan; The National Center for Diabetes, Endocrinology and Genetics, The University of Jordan, Amman, Jordan; Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Takehiro Iwata
- Department of Urology, Medical University of Vienna, Vienna, Austria; Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shoji Kimura
- Department of Urology, Medical University of Vienna, Vienna, Austria; Department of Urology, Jikei University School of Medicine, Tokyo, Japan
| | - Anoud Haddad
- Department of Special Surgery, Jordan University Hospital, The University of Jordan, Amman, Jordan
| | - Hashim Al-Ani
- Department of Special Surgery, Jordan University Hospital, The University of Jordan, Amman, Jordan
| | - Lana Abusubaih
- Department of Special Surgery, Jordan University Hospital, The University of Jordan, Amman, Jordan
| | - Marco Moschini
- Department of Urology, Medical University of Vienna, Vienna, Austria; Klinik für Urologie, Luzerner Kantonsspital, Lucerne, Switzerland
| | - Alberto Briganti
- Division of Oncology, Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Shahrokh F Shariat
- Department of Special Surgery, Jordan University Hospital, The University of Jordan, Amman, Jordan; Department of Urology, Medical University of Vienna, Vienna, Austria; Department of Urology, Weill Cornell Medical College, New York, NY, USA; Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Karl Landsteiner Institute of Urology and Andrology, Vienna, Austria; Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia.
| |
Collapse
|
15
|
Hashimoto T, Nakashima J, Kashima T, Yamaguchi Y, Satake N, Nakagami Y, Namiki K, Ohno Y. Predicting factors for progression to castration resistance prostate cancer after biochemical recurrence in patients with clinically localized prostate cancer who underwent radical prostatectomy. Int J Clin Oncol 2020; 25:1704-1710. [PMID: 32500468 DOI: 10.1007/s10147-020-01716-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 05/25/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND To determine prognostic factors associated with progression to castration-resistant prostate cancer following biochemical recurrence which is lethal prostate cancer and establish a risk stratification model of progression to castration-resistant prostate cancer. METHODS We retrospectively reviewed the data of 550 patients who experienced biochemical recurrence after radical prostatectomy. The endpoint of the present study was progression to castration-resistant prostate cancer. The actuarial probabilities of progression to castration-resistant prostate cancer-free survival were determined using Kaplan-Meier analysis. Univariate and multivariate Cox proportional hazards regression analyses were used to identify independent predictors of biochemical recurrence. RESULTS Fifty-two patients experienced progression to castration-resistant prostate cancer during the follow-up period. The progression to castration-resistant prostate cancer-free survival rate after biochemical recurrence at 10 years was 76.8%. In multivariate analysis, pathological Gleason score ≥ 9, lymphovascular invasion, and prostate-specific antigen velocity ≥ 0.4 ng/mL/year were independent predictive factors for progression to castration-resistant prostate cancer. The patients were stratified into three groups using a risk stratification model incorporating these variables. The 10-year progression to castration-resistant prostate cancer-free survival rates were 96.7% in the low-risk group, 84.7% in the intermediate-risk group, and 24.5% in the high-risk group. CONCLUSIONS The present results suggest that the pathological Gleason score, lymphovascular invasion, and prostate-specific antigen velocity were independent predictive factors for progression to castration-resistant prostate cancer. The risk stratification model established in the present study could be useful for patient counseling and in identifying patients with a poor prognosis.
Collapse
Affiliation(s)
- Takeshi Hashimoto
- Department of Urology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan.
| | - Jun Nakashima
- Department of Urology, Sanno Hospital, Tokyo, Japan.,Clinical Medicine Research Center, International University of Health and Welfare, Tokyo, Japan
| | - Takeshi Kashima
- Department of Urology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Yuri Yamaguchi
- Department of Urology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Naoya Satake
- Department of Urology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Yoshihiro Nakagami
- Department of Urology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Kazunori Namiki
- Department of Urology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Yoshio Ohno
- Department of Urology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| |
Collapse
|
16
|
Longitudinal model-based meta-analysis for survival probabilities in patients with castration-resistant prostate cancer. Eur J Clin Pharmacol 2020; 76:589-601. [PMID: 31925454 DOI: 10.1007/s00228-020-02829-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 01/03/2020] [Indexed: 01/11/2023]
Abstract
PURPOSE The aims of this longitudinal model-based meta-analysis (MBMA) were to indirectly compare the time courses of survival probabilities and to identify corresponding potential significant covariates across approved drugs in patients with castration-resistant prostate cancer (CRPC). METHODS A systematic literature review for monotherapy studies in patients with CRPC was conducted up to August 8, 2018. The time courses of progression-free survival (PFS) and overall survival (OS) were fitted with parametric survival models. Covariate analyses were performed to determine the impact of treatment drugs, dosing regimens, and patient characteristics on the survival probabilities. Simulations were carried out to quantify the magnitude of covariate effects. RESULTS A total of 146 studies including clinical trials and real-world data on longitudinal survival probabilities in 20,712 patients with CRPC were included in our meta-database. The time courses of PFS and OS probabilities were best described by the log-logistic model. There was no significant difference in median OS and PFS between docetaxel, cabazitaxel, abiraterone acetate, and enzalutamide. There was no significant dose-response relationship in PFS or OS for docetaxel at 50 to 120 mg/m2 every 3 weeks (Q3W) and cabazitaxel at 20 to 25 mg/m2 Q3W. Model-based simulations indicated that PFS probability was associated with chemotherapy, Gleason score, and baseline prostate-specific antigen (BLPSA), while OS probability was associated with chemotherapy, Gleason score, visceral metastasis, Eastern Cooperative Oncology Group performance status, and BLPSA. CONCLUSION Our modeling and simulation framework can be applied to support indirect comparison, dose selection, and go/no-go decision-making for new agents targeting CRPC.
Collapse
|
17
|
Smith MR, Mehra M, Nair S, Lawson J, Small EJ. Relationship Between Metastasis-free Survival and Overall Survival in Patients With Nonmetastatic Castration-resistant Prostate Cancer. Clin Genitourin Cancer 2019; 18:e180-e189. [PMID: 31980408 DOI: 10.1016/j.clgc.2019.10.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 10/28/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Metastasis-free survival (MFS) has been shown to be predictive of overall survival (OS) in hormone-sensitive localized prostate cancer. We evaluated the relationship between MFS and OS in nonmetastatic castration-resistant prostate cancer (nmCRPC). PATIENTS AND METHODS A retrospective analysis of 1207 high-risk patients with nmCRPC from the SPARTAN study (clinicaltrials.gov, NCT01946204) was undertaken. Landmark analyses of MFS status at several time points from randomization were performed to minimize guarantee-time bias. Hazard ratio (HR) of death as a function of MFS status was estimated based on a Cox proportional hazards model with 2-sided 95% confidence interval (CI). Estimated HRs were adjusted for stratification factors. Correlation analysis was performed using the Fleischer method. RESULTS At all time points, MFS status strongly predicted OS. At landmark time points of 6, 9, and 12 months, risk of death was significantly higher for patients with metastases versus those without (adjusted HR at 6 months = 4.12; 95% CI, 2.60-6.54; P < .0001). MFS was positively correlated with OS based on the Fleischer method (HR, 0.69; 95% CI, 0.69-0.70; P < .0001). Approximately one-half of the variability in OS can be explained by MFS. CONCLUSION Metastasis development, regardless of time point, is associated with significantly greater risk of death in men with high-risk nmCRPC; hence, MFS is predictive of OS.
Collapse
Affiliation(s)
- Matthew R Smith
- Department of Hematology and Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA.
| | - Maneesha Mehra
- Health Economics Design and Analytics, Janssen Research & Development, Raritan, NJ
| | - Sandhya Nair
- Heath Economics Design and Analytics, Janssen Research & Development, Beerse, Belgium
| | - Joe Lawson
- Health Economics Design and Analytics, Janssen Research & Development, Raritan, NJ
| | - Eric J Small
- Division of Hematology and Oncology, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA
| |
Collapse
|
18
|
Chen W, Li L, Ji S, Song X, Lu W, Zhou T. Evaluation of potential surrogate endpoints for prediction of overall survival in patients with castration-resistant prostate cancer: trial-level meta-analysis. Eur J Clin Pharmacol 2019; 75:1521-1532. [DOI: 10.1007/s00228-019-02736-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 08/09/2019] [Indexed: 01/25/2023]
|
19
|
Armstrong AJ, Szmulewitz RZ, Petrylak DP, Holzbeierlein J, Villers A, Azad A, Alcaraz A, Alekseev B, Iguchi T, Shore ND, Rosbrook B, Sugg J, Baron B, Chen L, Stenzl A. ARCHES: A Randomized, Phase III Study of Androgen Deprivation Therapy With Enzalutamide or Placebo in Men With Metastatic Hormone-Sensitive Prostate Cancer. J Clin Oncol 2019; 37:2974-2986. [PMID: 31329516 PMCID: PMC6839905 DOI: 10.1200/jco.19.00799] [Citation(s) in RCA: 725] [Impact Index Per Article: 120.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Enzalutamide, a potent androgen-receptor inhibitor, has demonstrated significant benefits in metastatic and nonmetastatic castration-resistant prostate cancer. We evaluated the efficacy and safety of enzalutamide in metastatic hormone-sensitive prostate cancer (mHSPC). METHODS ARCHES (ClinicalTrials.gov identifier: NCT02677896) is a multinational, double-blind, phase III trial, wherein 1,150 men with mHSPC were randomly assigned 1:1 to enzalutamide (160 mg/day) or placebo, plus androgen deprivation therapy (ADT), stratified by disease volume and prior docetaxel chemotherapy. The primary end point was radiographic progression-free survival. RESULTS As of October 14, 2018, the risk of radiographic progression or death was significantly reduced with enzalutamide plus ADT versus placebo plus ADT (hazard ratio, 0.39; 95% CI, 0.30 to 0.50; P < .001; median not reached v 19.0 months). Similar significant improvements in radiographic progression-free survival were reported in prespecified subgroups on the basis of disease volume and prior docetaxel therapy. Enzalutamide plus ADT significantly reduced the risk of prostate-specific antigen progression, initiation of new antineoplastic therapy, first symptomatic skeletal event, castration resistance, and reduced risk of pain progression. More men achieved an undetectable prostate-specific antigen level and/or an objective response with enzalutamide plus ADT (P < .001). Patients in both treatment groups reported a high baseline level of quality of life, which was maintained over time. Grade 3 or greater adverse events were reported in 24.3% of patients who received enzalutamide plus ADT versus 25.6% of patients who received placebo plus ADT, with no unexpected adverse events. CONCLUSION Enzalutamide with ADT significantly reduced the risk of metastatic progression or death over time versus placebo plus ADT in men with mHSPC, including those with low-volume disease and/or prior docetaxel, with a safety analysis that seems consistent with the safety profile of enzalutamide in previous clinical trials in castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Andrew J Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC
| | | | | | | | | | - Arun Azad
- Monash Health, Melbourne, Victoria, Australia
| | | | - Boris Alekseev
- Hertzen Moscow Cancer Research Institute, Moscow, Russia
| | - Taro Iguchi
- Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Neal D Shore
- Carolina Urologic Research Center, Myrtle Beach, SC
| | | | | | | | | | - Arnulf Stenzl
- Eberhard Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
20
|
Clinical Trials and Their Principles in Urologic Oncology. Urol Oncol 2019. [DOI: 10.1007/978-3-319-42623-5_54] [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]
|
21
|
Kashiwabara T, Suda S. Usefulness of combined androgen blockade therapy with gonadotropin-releasing hormone antagonist for bone metastatic prostate cancer with pretreatment prostate-specific antigen level ≥ 50 ng/mL. BMC Cancer 2018; 18:619. [PMID: 29855278 PMCID: PMC5984333 DOI: 10.1186/s12885-018-4541-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 05/21/2018] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND This study was performed to examine the usefulness of combined androgen blockade (CAB) therapy with a gonadotropin-releasing hormone (GnRH) antagonist (CAB-antagonist therapy), instead of CAB therapy with GnRH agonist (CAB-agonist therapy) against very high-risk prostate cancer (Pca). METHODS We retrospectively studied 84 Pca patients with pretreatment prostate-specific antigen (PSA) level ≥ 50 ng/mL, who were pathologically diagnosed between January 2007 and December 2016. GnRH antagonist was administered to 34 patients and GnRH agonist was administered to 50 patients. All patients received concurrent antiandrogen treatment. The primary end point was PSA progression-free survival (PSA-PFS). RESULTS PSA-PFS was significantly longer for the CAB-antagonist group compared to the CAB-agonist group (log-rank test, P < 0.01) in Pca patients with more than six bone metastases (the extent of disease [EOD] grade 2-4). On multivariate analysis, CAB-antagonist therapy was shown to be a possible prognostic factor for PSA-PFS (adjusted hazard ratio: 0.41, 95% confidence interval: 0.16-0.90, P = 0.03). CONCLUSIONS CAB-antagonist therapy may be a useful option in bone metastatic Pca patients with EOD grade 2-4.
Collapse
Affiliation(s)
- Takeshi Kashiwabara
- Department of Urology, Saku Central Hospital, 197 Usuda, Saku, Nagano, 384-0393 Japan
| | - Sayo Suda
- Department of Urology, Saku Central Hospital, 197 Usuda, Saku, Nagano, 384-0393 Japan
| |
Collapse
|
22
|
Suciu S, Eggermont AMM, Lorigan P, Kirkwood JM, Markovic SN, Garbe C, Cameron D, Kotapati S, Chen TT, Wheatley K, Ives N, de Schaetzen G, Efendi A, Buyse M. Relapse-Free Survival as a Surrogate for Overall Survival in the Evaluation of Stage II-III Melanoma Adjuvant Therapy. J Natl Cancer Inst 2018; 110:4091329. [PMID: 28922786 DOI: 10.1093/jnci/djx133] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 05/26/2017] [Indexed: 02/11/2024] Open
Abstract
Background We assessed whether relapse-free survival (RFS; time until recurrence/death) is a valid surrogate for overall survival (OS) among resected stage II-III melanoma patients through a meta-analysis of randomized controlled trials. Methods Individual patient data (IPD) on RFS and OS were collected from 5826 patients enrolled in 11 randomized adjuvant trials comparing interferon (IFN) to observation. In addition, IPD from two studies comparing IFN and vaccination in 989 patients were included. A two-level modeling approach was used for assessing Spearman's patient-level correlation (rho) of RFS and OS and the trial-level coefficient of determination (R²) of the treatment effects on RFS and on OS. The results were validated externally in 13 adjuvant studies without available IPD. We then tested the results on the European Organisation for Research and Treatment of Cancer (EORTC) 18071 double-blind trial comparing ipilimumab 10 mg/kg with placebo, which showed a statistically significant impact of the checkpoint inhibitor on RFS and OS. All statistical tests were two-sided. Results With a median follow-up of seven years, 12 of 13 trials showed a consistency between the IFN vs No IFN differences regarding RFS (hazard ratio [HR]RFS = 0.88) and OS (HROS = 0.91), but the small trial, Eastern Cooperative Oncology Group 2696, was an outlier (HRRFS = 0.72 vs HROS = 1.11). Therefore, even if rho was high, R² was low and could not reliably be estimated. Based on the 12 trials, rho remained high (0.89), and the hazard ratios for RFS and OS were strongly correlated (R² = 0.91). The surrogate threshold effect for RFS was estimated to be 0.77. For the EORTC 18071 trial, the hazard ratio for RFS was 0.75, predicting an effect of ipilimumab on OS. This was subsequently confirmed (HROS = 0.72, 95.1% confidence interval = 0.58 to 0.88, P = .001). Conclusions In high-risk stage II-III melanoma, RFS appeared to be a valid surrogate end point for OS for adjuvant randomized studies assessing interferon or a checkpoint inhibitor. In future similar adjuvant studies, a hazard ratio for RFS of 0.77 or less would predict a treatment impact on OS.
Collapse
Affiliation(s)
- Stefan Suciu
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium; Gustave Roussy Cancer Campus Grand Paris, Villejuif, France; The Christie NHS Foundation Trust, Manchester, UK; University of Pittsburgh Cancer Institute and School of Medicine, Pittsburgh, PA; Mayo Clinic Rochester, Rochester, MN; University of Tubingen, Tubingen, Germany; University of Edinburgh, Western General Hospital, Edinburgh, UK; Bristol-Myers Squibb, Wallingford, CT; Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK; Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK; Universitas Brawijaya, Malang, Indonesia; IDDI, Louvain-la-Neuve, Belgium
| | - Alexander M M Eggermont
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium; Gustave Roussy Cancer Campus Grand Paris, Villejuif, France; The Christie NHS Foundation Trust, Manchester, UK; University of Pittsburgh Cancer Institute and School of Medicine, Pittsburgh, PA; Mayo Clinic Rochester, Rochester, MN; University of Tubingen, Tubingen, Germany; University of Edinburgh, Western General Hospital, Edinburgh, UK; Bristol-Myers Squibb, Wallingford, CT; Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK; Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK; Universitas Brawijaya, Malang, Indonesia; IDDI, Louvain-la-Neuve, Belgium
| | - Paul Lorigan
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium; Gustave Roussy Cancer Campus Grand Paris, Villejuif, France; The Christie NHS Foundation Trust, Manchester, UK; University of Pittsburgh Cancer Institute and School of Medicine, Pittsburgh, PA; Mayo Clinic Rochester, Rochester, MN; University of Tubingen, Tubingen, Germany; University of Edinburgh, Western General Hospital, Edinburgh, UK; Bristol-Myers Squibb, Wallingford, CT; Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK; Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK; Universitas Brawijaya, Malang, Indonesia; IDDI, Louvain-la-Neuve, Belgium
| | - John M Kirkwood
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium; Gustave Roussy Cancer Campus Grand Paris, Villejuif, France; The Christie NHS Foundation Trust, Manchester, UK; University of Pittsburgh Cancer Institute and School of Medicine, Pittsburgh, PA; Mayo Clinic Rochester, Rochester, MN; University of Tubingen, Tubingen, Germany; University of Edinburgh, Western General Hospital, Edinburgh, UK; Bristol-Myers Squibb, Wallingford, CT; Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK; Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK; Universitas Brawijaya, Malang, Indonesia; IDDI, Louvain-la-Neuve, Belgium
| | - Svetomir N Markovic
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium; Gustave Roussy Cancer Campus Grand Paris, Villejuif, France; The Christie NHS Foundation Trust, Manchester, UK; University of Pittsburgh Cancer Institute and School of Medicine, Pittsburgh, PA; Mayo Clinic Rochester, Rochester, MN; University of Tubingen, Tubingen, Germany; University of Edinburgh, Western General Hospital, Edinburgh, UK; Bristol-Myers Squibb, Wallingford, CT; Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK; Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK; Universitas Brawijaya, Malang, Indonesia; IDDI, Louvain-la-Neuve, Belgium
| | - Claus Garbe
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium; Gustave Roussy Cancer Campus Grand Paris, Villejuif, France; The Christie NHS Foundation Trust, Manchester, UK; University of Pittsburgh Cancer Institute and School of Medicine, Pittsburgh, PA; Mayo Clinic Rochester, Rochester, MN; University of Tubingen, Tubingen, Germany; University of Edinburgh, Western General Hospital, Edinburgh, UK; Bristol-Myers Squibb, Wallingford, CT; Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK; Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK; Universitas Brawijaya, Malang, Indonesia; IDDI, Louvain-la-Neuve, Belgium
| | - David Cameron
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium; Gustave Roussy Cancer Campus Grand Paris, Villejuif, France; The Christie NHS Foundation Trust, Manchester, UK; University of Pittsburgh Cancer Institute and School of Medicine, Pittsburgh, PA; Mayo Clinic Rochester, Rochester, MN; University of Tubingen, Tubingen, Germany; University of Edinburgh, Western General Hospital, Edinburgh, UK; Bristol-Myers Squibb, Wallingford, CT; Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK; Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK; Universitas Brawijaya, Malang, Indonesia; IDDI, Louvain-la-Neuve, Belgium
| | - Srividya Kotapati
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium; Gustave Roussy Cancer Campus Grand Paris, Villejuif, France; The Christie NHS Foundation Trust, Manchester, UK; University of Pittsburgh Cancer Institute and School of Medicine, Pittsburgh, PA; Mayo Clinic Rochester, Rochester, MN; University of Tubingen, Tubingen, Germany; University of Edinburgh, Western General Hospital, Edinburgh, UK; Bristol-Myers Squibb, Wallingford, CT; Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK; Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK; Universitas Brawijaya, Malang, Indonesia; IDDI, Louvain-la-Neuve, Belgium
| | - Tai-Tsang Chen
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium; Gustave Roussy Cancer Campus Grand Paris, Villejuif, France; The Christie NHS Foundation Trust, Manchester, UK; University of Pittsburgh Cancer Institute and School of Medicine, Pittsburgh, PA; Mayo Clinic Rochester, Rochester, MN; University of Tubingen, Tubingen, Germany; University of Edinburgh, Western General Hospital, Edinburgh, UK; Bristol-Myers Squibb, Wallingford, CT; Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK; Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK; Universitas Brawijaya, Malang, Indonesia; IDDI, Louvain-la-Neuve, Belgium
| | - Keith Wheatley
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium; Gustave Roussy Cancer Campus Grand Paris, Villejuif, France; The Christie NHS Foundation Trust, Manchester, UK; University of Pittsburgh Cancer Institute and School of Medicine, Pittsburgh, PA; Mayo Clinic Rochester, Rochester, MN; University of Tubingen, Tubingen, Germany; University of Edinburgh, Western General Hospital, Edinburgh, UK; Bristol-Myers Squibb, Wallingford, CT; Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK; Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK; Universitas Brawijaya, Malang, Indonesia; IDDI, Louvain-la-Neuve, Belgium
| | - Natalie Ives
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium; Gustave Roussy Cancer Campus Grand Paris, Villejuif, France; The Christie NHS Foundation Trust, Manchester, UK; University of Pittsburgh Cancer Institute and School of Medicine, Pittsburgh, PA; Mayo Clinic Rochester, Rochester, MN; University of Tubingen, Tubingen, Germany; University of Edinburgh, Western General Hospital, Edinburgh, UK; Bristol-Myers Squibb, Wallingford, CT; Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK; Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK; Universitas Brawijaya, Malang, Indonesia; IDDI, Louvain-la-Neuve, Belgium
| | - Gaetan de Schaetzen
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium; Gustave Roussy Cancer Campus Grand Paris, Villejuif, France; The Christie NHS Foundation Trust, Manchester, UK; University of Pittsburgh Cancer Institute and School of Medicine, Pittsburgh, PA; Mayo Clinic Rochester, Rochester, MN; University of Tubingen, Tubingen, Germany; University of Edinburgh, Western General Hospital, Edinburgh, UK; Bristol-Myers Squibb, Wallingford, CT; Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK; Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK; Universitas Brawijaya, Malang, Indonesia; IDDI, Louvain-la-Neuve, Belgium
| | - Achmad Efendi
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium; Gustave Roussy Cancer Campus Grand Paris, Villejuif, France; The Christie NHS Foundation Trust, Manchester, UK; University of Pittsburgh Cancer Institute and School of Medicine, Pittsburgh, PA; Mayo Clinic Rochester, Rochester, MN; University of Tubingen, Tubingen, Germany; University of Edinburgh, Western General Hospital, Edinburgh, UK; Bristol-Myers Squibb, Wallingford, CT; Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK; Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK; Universitas Brawijaya, Malang, Indonesia; IDDI, Louvain-la-Neuve, Belgium
| | - Marc Buyse
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium; Gustave Roussy Cancer Campus Grand Paris, Villejuif, France; The Christie NHS Foundation Trust, Manchester, UK; University of Pittsburgh Cancer Institute and School of Medicine, Pittsburgh, PA; Mayo Clinic Rochester, Rochester, MN; University of Tubingen, Tubingen, Germany; University of Edinburgh, Western General Hospital, Edinburgh, UK; Bristol-Myers Squibb, Wallingford, CT; Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK; Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK; Universitas Brawijaya, Malang, Indonesia; IDDI, Louvain-la-Neuve, Belgium
| |
Collapse
|
23
|
Li D, Lv H, Hao X, Dong Y, Dai H, Song Y. Prognostic value of bone scan index as an imaging biomarker in metastatic prostate cancer: a meta-analysis. Oncotarget 2017; 8:84449-84458. [PMID: 29137438 PMCID: PMC5663610 DOI: 10.18632/oncotarget.19680] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 06/30/2017] [Indexed: 12/27/2022] Open
Abstract
Background The prognostic value of the bone scan index (BSI) in metastatic prostate cancer (mPCa) remained controversial. Therefore, we performed a meta-analysis to determine the predictive value of BSI and survival in patients with mPCa. Materials and Methods A literature search was performed in PubMed, Embase, Web of Science and Cochrane library databases. Hazard ratios (HRs), concordance indices (C-indices) were extracted to estimate the relationship between BSI and survival in patients with mPCa. Subgroup analyses were conducted on different types of mPCa, ethnics, cut-off values and sample sizes. Results 14 high quality studies involving 1295 patients with mPCa were included in this meta-analysis. The pooled results indicated that high basline BSI and elevated BSI change on treatment (ΔBSI) were significantly predictive of poor overall survial (HR = 1.29, P < 0.001; HR = 1.27, P < 0.001, respectively). Baseline BSI was also significantly related to cancer specific survival (HR = 1.65, P = 0.019) and prostate specific antigen recurrence survival (HR = 2.26, P < 0.001). Subgroup analysis supported main results. Moreover, BSI could increase the C-indices of predictive models. Conclusions Baseline BSI and ΔBSI may be beneficial to mPCa prognosis in clinical monitor and treatment. Further high quality studies with larger sample size are required in the future.
Collapse
Affiliation(s)
- Dongyang Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Hang Lv
- Department of Urology, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, Liaoning 110042, P.R. China
| | - Xuanyu Hao
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Yudi Dong
- Department of Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Huixu Dai
- Department of Clinical Epidemiology and Evidence-based Medicine, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yongsheng Song
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
24
|
Brookman-May SD, Mir MC, May M, Klatte T. Clinical Trials and Their Principles in Urologic Oncology. Urol Oncol 2017. [DOI: 10.1007/978-3-319-42603-7_54-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
25
|
Trial Level Analysis of Prostate-Specific Antigen-Related Versus Unrelated Endpoints in Phase III Trials of First-Line and Second-Line Medical Treatments of Patients With Metastatic Castration-Resistant Prostate Cancer. Clin Genitourin Cancer 2016; 14:389-397. [DOI: 10.1016/j.clgc.2016.03.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 03/07/2016] [Accepted: 03/19/2016] [Indexed: 11/21/2022]
|
26
|
Validation of surrogate endpoints in advanced solid tumors: systematic review of statistical methods, results, and implications for policy makers. Int J Technol Assess Health Care 2016; 30:312-24. [PMID: 25308694 DOI: 10.1017/s0266462314000300] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Licensing of, and coverage decisions on, new therapies should rely on evidence from patient-relevant endpoints such as overall survival (OS). Nevertheless, evidence from surrogate endpoints may also be useful, as it may not only expedite the regulatory approval of new therapies but also inform coverage decisions. It is, therefore, essential that candidate surrogate endpoints be properly validated. However, there is no consensus on statistical methods for such validation and on how the evidence thus derived should be applied by policy makers. METHODS We review current statistical approaches to surrogate-endpoint validation based on meta-analysis in various advanced-tumor settings. We assessed the suitability of two surrogates (progression-free survival [PFS] and time-to-progression [TTP]) using three current validation frameworks: Elston and Taylor's framework, the German Institute of Quality and Efficiency in Health Care's (IQWiG) framework and the Biomarker-Surrogacy Evaluation Schema (BSES3). RESULTS A wide variety of statistical methods have been used to assess surrogacy. The strength of the association between the two surrogates and OS was generally low. The level of evidence (observation-level versus treatment-level) available varied considerably by cancer type, by evaluation tools and was not always consistent even within one specific cancer type. CONCLUSIONS Not in all solid tumors the treatment-level association between PFS or TTP and OS has been investigated. According to IQWiG's framework, only PFS achieved acceptable evidence of surrogacy in metastatic colorectal and ovarian cancer treated with cytotoxic agents. Our study emphasizes the challenges of surrogate-endpoint validation and the importance of building consensus on the development of evaluation frameworks.
Collapse
|
27
|
Maroto P, Solsona E, Gallardo E, Mellado B, Morote J, Arranz JÁ, Gómez-Veiga F, Unda M, Climent MÁ, Alcaraz A. Expert opinion on first-line therapy in the treatment of castration-resistant prostate cancer. Crit Rev Oncol Hematol 2016; 100:127-36. [PMID: 26363809 DOI: 10.1016/j.critrevonc.2015.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 06/26/2015] [Accepted: 07/28/2015] [Indexed: 10/23/2022] Open
Abstract
Treatment of metastatic castration-resistant prostate cancer (mCRPC) has been revolutionized in recent years. It is well known that androgen receptor is still active in most patients with disease progression and serum testosterone levels <50 ng/dL. Moreover, further hormonal maneuvers, either through decreasing androgen levels (abiraterone) or by targeting the androgen receptor (AR) pathway (enzalutamide), prolong survival. In addition, a new cytostatic able to overcome docetaxel resistance, cabazitaxel, and the radioisotope radium 223 have been incorporated to the armamentarium of mCRPC. mCRPC is not only a heterogeneous tumor, it changes over time developing neuroendocrine features or selection of clones resistant to hormonal maneuvers. In addition, the multiplicity of current treatments, make it necessary to design algorithms that help the specialist to choose the most appropriate treatment for a particular patient. The lack of randomized trials comparing face to face the different available options limit the scope of this review. In this article, the authors describe the prognostic factors for first line therapy in patients with mCRPC, and propose a treatment algorithm for mCRPC based on the levels of scientific evidence available and, if not available, on the consensus between medical professionals. Finally, the panel discuss how to define progressive disease in the setting of mCRPC and treatment with targeted therapies.
Collapse
Affiliation(s)
- Pablo Maroto
- Department of Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.
| | - Eduardo Solsona
- Department of Urology, Instituto Valenciano de Oncología, Valencia, Spain
| | - Enrique Gallardo
- Department of Oncology, Parc Taulí Sabadell Hospital Universitari, Sabadell, Barcelona, Spain
| | - Begoña Mellado
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | - Juan Morote
- Department of Urology, Hospital Vall d́Hebrón, Barcelona, Spain
| | - José Ángel Arranz
- Department of Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | | - Miguel Unda
- Department of Urology, Hospital de Basurto, Bilbao, Spain
| | | | | |
Collapse
|
28
|
Scher HI, Morris MJ, Stadler WM, Higano C, Basch E, Fizazi K, Antonarakis ES, Beer TM, Carducci MA, Chi KN, Corn PG, de Bono JS, Dreicer R, George DJ, Heath EI, Hussain M, Kelly WK, Liu G, Logothetis C, Nanus D, Stein MN, Rathkopf DE, Slovin SF, Ryan CJ, Sartor O, Small EJ, Smith MR, Sternberg CN, Taplin ME, Wilding G, Nelson PS, Schwartz LH, Halabi S, Kantoff PW, Armstrong AJ. Trial Design and Objectives for Castration-Resistant Prostate Cancer: Updated Recommendations From the Prostate Cancer Clinical Trials Working Group 3. J Clin Oncol 2016; 34:1402-18. [PMID: 26903579 DOI: 10.1200/jco.2015.64.2702] [Citation(s) in RCA: 1187] [Impact Index Per Article: 131.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Evolving treatments, disease phenotypes, and biology, together with a changing drug development environment, have created the need to revise castration-resistant prostate cancer (CRPC) clinical trial recommendations to succeed those from prior Prostate Cancer Clinical Trials Working Groups. METHODS An international expert committee of prostate cancer clinical investigators (the Prostate Cancer Clinical Trials Working Group 3 [PCWG3]) was reconvened and expanded and met in 2012-2015 to formulate updated criteria on the basis of emerging trial data and validation studies of the Prostate Cancer Clinical Trials Working Group 2 recommendations. RESULTS PCWG3 recommends that baseline patient assessment include tumor histology, detailed records of prior systemic treatments and responses, and a detailed reporting of disease subtypes based on an anatomic pattern of metastatic spread. New recommendations for trial outcome measures include the time to event end point of symptomatic skeletal events, as well as time to first metastasis and time to progression for trials in the nonmetastatic CRPC state. PCWG3 introduces the concept of no longer clinically benefiting to underscore the distinction between first evidence of progression and the clinical need to terminate or change treatment, and the importance of documenting progression in existing lesions as distinct from the development of new lesions. Serial biologic profiling using tumor samples from biopsies, blood-based diagnostics, and/or imaging is also recommended to gain insight into mechanisms of resistance and to identify predictive biomarkers of sensitivity for use in prospective trials. CONCLUSION PCWG3 moves drug development closer to unmet needs in clinical practice by focusing on disease manifestations most likely to affect prognosis adversely for therapeutics tested in both nonmetastatic and metastatic CRPC populations. Consultation with regulatory authorities is recommended if a trial is intended to seek support for drug approval.
Collapse
Affiliation(s)
- Howard I Scher
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Michael J Morris
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Walter M Stadler
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Celestia Higano
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Ethan Basch
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Karim Fizazi
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Emmanuel S Antonarakis
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Tomasz M Beer
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Michael A Carducci
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Kim N Chi
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Paul G Corn
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Johann S de Bono
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Robert Dreicer
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Daniel J George
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Elisabeth I Heath
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Maha Hussain
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Wm Kevin Kelly
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Glenn Liu
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Christopher Logothetis
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - David Nanus
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Mark N Stein
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Dana E Rathkopf
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Susan F Slovin
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Charles J Ryan
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Oliver Sartor
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Eric J Small
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Matthew Raymond Smith
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Cora N Sternberg
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Mary-Ellen Taplin
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - George Wilding
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Peter S Nelson
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Lawrence H Schwartz
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Susan Halabi
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Philip W Kantoff
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | - Andrew J Armstrong
- Howard I. Scher, Michael J. Morris, Dana E. Rathkopf, and Susan F. Slovin, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College; David Nanus, NewYork Presbyterian Weill Cornell Medical Center; Lawrence W. Schwartz, NewYork Presbyterian Columbia University Medical Center, New York, NY; Walter M. Stadler, University of Chicago Medicine, Chicago, IL; Celestina Higano and Peter S. Nelson, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA; Ethan Basch, University of North Carolina at Chapel Hill, Chapel Hill, NC; Emmanual S. Antonarakis and Michael A. Carducci, Johns Hopkins University School of Medicine, Baltimore, MD; Tomasz M. Beer, Oregon Health and Science University, Portland, OR; Paul G. Corn and Christopher Logothetis, MD Anderson Cancer Center, Houston, TX; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Daniel J. George, Susan Halabi, and Andrew J. Armstrong, Duke University and Duke Cancer Institute, Durham, NC; Elisabeth I. Heath, Wayne State University Karmanos Cancer Institute, Detroit; and Maha Hussain, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; Wm. Kevin Kelly, Sidney Kimmel School of Medicine at Thomas Jefferson University, Philadelphia, PA; Glenn Liu and George Wilding, University of Wisconsin Carbone Cancer Center, Madison, WI; Mark N. Stein, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ; Charles S. Ryan and Eric J. Small, University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Oliver Sartor, Tulane School of Medicine, New Orleans, LA; Matthew Raymond Smith, Massachusetts General Hospital Cancer Center and Harvard Medical School; Mary-Ellen Taplin and Philip W. Kantoff, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, Franc
| | | |
Collapse
|
29
|
Miyoshi Y, Yoneyama S, Kawahara T, Hattori Y, Teranishi JI, Kondo K, Moriyama M, Takebayashi S, Yokomizo Y, Yao M, Uemura H, Noguchi K. Prognostic value of the bone scan index using a computer-aided diagnosis system for bone scans in hormone-naive prostate cancer patients with bone metastases. BMC Cancer 2016; 16:128. [PMID: 26896160 PMCID: PMC4759962 DOI: 10.1186/s12885-016-2176-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 02/15/2016] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The bone scan index (BSI) using a computer-aided diagnosis system for bone scans is expected to be an objective and quantitative clinical tool for evaluating bone metastatic prostate cancer. This study aimed to evaluate the pretreatment BSI as a prognostic factor in hormone-naive prostate cancer patients with bone metastases. METHODS The study included 60 patients with hormone-naive, bone metastatic prostate cancer that was initially treated with combined androgen blockade therapy. The BONENAVI system was used for calculating the BSI. We evaluated the correlation between overall survival (OS) and pretreatment clinicopathological characteristics, including patients' age, initial prostate-specific antigen (PSA) value, Gleason scores, clinical TNM stage, and the BSI. Cox proportional hazards regression models were used for statistical analysis. RESULTS The median follow-up duration was 21.4 months. Clinical or PSA progression occurred in 37 (61.7%) patients and 18 (30.0%) received docetaxel. Death occurred in 16 (26.7%) patients. Of these deaths, 15 (25.0%) were due to prostate cancer. The median OS was not reached. In multivariate analysis, age and the BSI were independent prognostic factors for OS. We evaluated the discriminatory ability of our models, including or excluding BSI by quantifying the C-index. The BSI improved the C-index from 0.751 to 0.801 for OS. Median OS was not reached in patients with a BSI ≤ 1.9 and median OS was 34.8 months in patients with a BSI >1.9 (p = 0.039). CONCLUSIONS The pretreatment BSI and patients' age are independent prognostic factors for patients with hormone-naive, bone metastatic prostate cancer.
Collapse
Affiliation(s)
- Yasuhide Miyoshi
- Department of Urology and Renal Transplantation, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan.
| | - Shuko Yoneyama
- Department of Urology and Renal Transplantation, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan.
| | - Takashi Kawahara
- Department of Urology and Renal Transplantation, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan.
| | - Yusuke Hattori
- Department of Urology and Renal Transplantation, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan.
| | - Jun-ichi Teranishi
- Department of Urology and Renal Transplantation, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan.
| | - Keiichi Kondo
- Department of Urology and Renal Transplantation, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan.
| | - Masatoshi Moriyama
- Department of Urology, Yokohama Municipal Citizen's Hospital, Yokohama, Japan.
| | - Shigeo Takebayashi
- Department of Radiology, Yokohama City University Medical Center, Yokohama, Japan.
| | - Yumiko Yokomizo
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Masahiro Yao
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Hiroji Uemura
- Department of Urology and Renal Transplantation, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan.
| | - Kazumi Noguchi
- Department of Urology and Renal Transplantation, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024, Japan.
| |
Collapse
|
30
|
Lee J, Sato MM, Coel MN, Lee KH, Kwee SA. Prediction of PSA Progression in Castration-Resistant Prostate Cancer Based on Treatment-Associated Change in Tumor Burden Quantified by 18F-Fluorocholine PET/CT. J Nucl Med 2016; 57:1058-64. [PMID: 26912444 DOI: 10.2967/jnumed.115.169177] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 01/26/2016] [Indexed: 01/19/2023] Open
Abstract
UNLABELLED Measurements of metabolically active tumor volume (MATV) can be applied to (18)F-fluorocholine PET/CT to quantify whole-body tumor burden. This study evaluated the serial application of these measurements as systemic treatment response markers and predictors of disease progression in patients with castration-resistant prostate cancer (CRPC). METHODS Forty-two patients completed sequential (18)F-fluorocholine PET/CT scans before and 1-3 mo after starting treatment for CRPC. Whole-body tumor segmentation was applied to determine net MATV from each scan. Changes in net MATV were evaluated as predictors of time to prostate-specific antigen (PSA) progression by Kaplan-Meier and proportional hazards regression analysis. RESULTS Treatments consisted of chemotherapy in 16 patients, antiandrogens in 19 patients, (223)Ra-dichloride in 5 patients, and sipuleucel-T in 2 patients. A significant MATV response (defined as a ≥30% decrease in net MATV) was observed in 20 patients on the basis of in-treatment PET/CT performed an average of 51 d (median, 49 d) into treatment. Significantly longer times to PSA progression were observed in patients who exhibited an MATV response (418 d vs. 116 d, P = 0.0067). MATV response was associated with a hazard ratio of 0.246 (P = 0.0113) for PSA progression, which remained significant when adjusted for treatment type. CONCLUSION Significant changes in whole-body tumor burden can be measured on (18)F-fluorocholine PET/CT over the course of contemporary treatments for CRPC. In this study, these changes were found to be predictive of PSA progression as a potential surrogate marker of treatment outcome. Because (18)F-fluorocholine PET/CT can also be used for localizing resistant tumors, this modality can potentially complement other measures of response in the precision management of advanced prostate cancer.
Collapse
Affiliation(s)
- Joohee Lee
- Oncology Research Program, Queen's Medical Center, Honolulu, Hawaii Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Miles M Sato
- Oncology Research Program, Queen's Medical Center, Honolulu, Hawaii
| | - Marc N Coel
- Hamamatsu/Queen's PET Imaging Center, Queen's Medical Center, Honolulu, Hawaii; and
| | - Kyung-Han Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sandi A Kwee
- Hamamatsu/Queen's PET Imaging Center, Queen's Medical Center, Honolulu, Hawaii; and Department of Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| |
Collapse
|
31
|
Murthy P, Kidwell KM, Schott AF, Merajver SD, Griggs JJ, Smerage JD, Van Poznak CH, Wicha MS, Hayes DF, Henry NL. Clinical predictors of long-term survival in HER2-positive metastatic breast cancer. Breast Cancer Res Treat 2016; 155:589-95. [PMID: 26875184 DOI: 10.1007/s10549-016-3705-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 02/05/2016] [Indexed: 12/29/2022]
Abstract
Prior to availability of anti-HER2 therapies, HER2-positive metastatic breast cancer (MBC) was associated with a poor prognosis. Prospective randomized trials have demonstrated survival benefit from anti-HER2 treatments. Anecdotal observations have suggested that a small but meaningful fraction of patients with HER2-positive MBC may be "exceptional responders" with long survival. We hypothesized that demographic and/or clinicopathologic characteristics can be identified to distinguish short-term from long-term survivors. A retrospective, single-institution review of 168 patients with HER2-positive MBC who received treatment with anti-HER2 therapy in the metastatic setting was performed. Cox proportional hazards analysis was used to assess factors associated with long-term survival. Median overall survival from the time of breast cancer recurrence was 3.9 years (95 % CI 3.4-5.2). From the time of diagnosis of MBC, 56 (33 %) survived for 5 or more years and 12 (7 %) survived more than 10 years. Of the 66 patients diagnosed with central nervous system metastases, 9 (14 %) survived more than 5 years following that diagnosis. Younger age at diagnosis, lower stage, hormone receptor positive status, and only having one organ involved at diagnosis were associated with longer survival. Four patients discontinued anti-HER2 therapy and are without evidence of progression of disease after a median 7.4 years (0.2-12.0) since stopping therapy. In a cohort of patients with HER2-positive MBC treated primarily with trastuzumab and lapatinib, 7 % of patients were "exceptional responders." Combining these clinical factors with molecular determinants of prolonged survival may provide insights for individualizing treatment selection.
Collapse
Affiliation(s)
- Pooja Murthy
- Internal Medicine/Hematology/Oncology, University of Michigan Medical School, 1500 East Medical Center Dr, Med Inn Bldg C450, Ann Arbor, MI, 48109-5843, USA
| | - Kelley M Kidwell
- Biostatistics, University of Michigan School of Public Health, 1415 Washington Heights, SPH II M2525, Ann Arbor, MI, 48109-2029, USA
| | - Anne F Schott
- Internal Medicine/Hematology/Oncology, University of Michigan Medical School, 1500 East Medical Center Dr, Med Inn Bldg C450, Ann Arbor, MI, 48109-5843, USA
| | - Sofia D Merajver
- Internal Medicine/Hematology/Oncology, University of Michigan Medical School, 1500 East Medical Center Dr, Med Inn Bldg C450, Ann Arbor, MI, 48109-5843, USA
| | - Jennifer J Griggs
- Internal Medicine/Hematology/Oncology, University of Michigan Medical School, 1500 East Medical Center Dr, Med Inn Bldg C450, Ann Arbor, MI, 48109-5843, USA
| | - Jeffrey D Smerage
- Internal Medicine/Hematology/Oncology, University of Michigan Medical School, 1500 East Medical Center Dr, Med Inn Bldg C450, Ann Arbor, MI, 48109-5843, USA
| | - Catherine H Van Poznak
- Internal Medicine/Hematology/Oncology, University of Michigan Medical School, 1500 East Medical Center Dr, Med Inn Bldg C450, Ann Arbor, MI, 48109-5843, USA
| | - Max S Wicha
- Internal Medicine/Hematology/Oncology, University of Michigan Medical School, 1500 East Medical Center Dr, Med Inn Bldg C450, Ann Arbor, MI, 48109-5843, USA
| | - Daniel F Hayes
- Internal Medicine/Hematology/Oncology, University of Michigan Medical School, 1500 East Medical Center Dr, Med Inn Bldg C450, Ann Arbor, MI, 48109-5843, USA
| | - N Lynn Henry
- Internal Medicine/Hematology/Oncology, University of Michigan Medical School, 1500 East Medical Center Dr, Med Inn Bldg C450, Ann Arbor, MI, 48109-5843, USA.
| |
Collapse
|
32
|
Ivanyi P, Koenig J, Trummer A, Busch JF, Seidel C, Reuter CW, Ganser A, Grünwald V. Does the onset of bone metastasis in sunitinib-treated renal cell carcinoma patients impact the overall survival? World J Urol 2015; 34:909-15. [PMID: 26586475 DOI: 10.1007/s00345-015-1707-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 10/06/2015] [Indexed: 12/24/2022] Open
Abstract
PURPOSE To evaluate the impact of bone metastasis (BM) onset toward prognosis in metastatic renal cell carcinoma (mRCC) patients treated with sunitinib. METHODS mRCC patients with BM and sunitinib as first targeted therapy between May 2005 and December 2012 were retrospectively analyzed. Patients with synchronous (s) BM or metachronous (m) BM were compared with regard to treatment and outcome [time to clinical progression (TTcP), overall survival (OS), skeletal-related events (SRE)]. Descriptive statistics, Kaplan-Meier estimation of TTcP and OS, Cox regression analyses, and a landmark analysis were administered. RESULTS BM was identified in 127 mRCC patients; thereof, 82 sunitinib-treated patients were analyzed [sBM n = 57 (69.5 %), mBM n = 25 (30.5 %)]. Higher tumor grading (p = 0.029), male predominance (p = 0.02), and less second-line therapy (p = 0.001) were detected in sBM compared to mBM. SRE remained similar between subgroups (p = 0.462). TTcP during sunitinib was similar [median sBM 8.1 (95 % CI 3.9-12.3) vs. mBM 8.7 (95 % CI 2.7-14.8) months, p = 0.903]. OS remained significantly inferior in sBM patients compared to mBM [median sBM 21.1 (95 % CI 16-26.2) months vs. mBM 38.5 (95 % CI 15-62) months, p = 0.001], which was confirmed by landmark analyses at 1.5, 3, 6, 9, and 12 months. However, OS after occurrence of BM was similar in both groups [median sBM 24.2 (95 % CI 17.3-31.1) months vs. mBM 17.2 (95 % CI 8.4-26) months, p = 0.519]. CONCLUSIONS mBM is associated with an improved OS compared to sBM in mRCC with sunitinib treatment, despite similar efficacy of sunitinib treatment in both groups of patients.
Collapse
Affiliation(s)
- P Ivanyi
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - J Koenig
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - A Trummer
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - J F Busch
- Clinic of Urology, University Hospital Berlin, Charité, Berlin, Germany
| | - C Seidel
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Medical Center Eppendorf, Hamburg, Germany
| | - C W Reuter
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - A Ganser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - V Grünwald
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
33
|
Davari M, Ashrafi F, Maracy M, Aslani A, Tabatabaei M. Cost-effectiveness Analysis of Cetuximab in Treatment of Metastatic Colorectal Cancer in Iranian Pharmaceutical Market. Int J Prev Med 2015; 6:63. [PMID: 26288707 PMCID: PMC4521302 DOI: 10.4103/2008-7802.161068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 03/19/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Cetuximab is a monoclonal antibody which acts against the epidermal growth-factor receptor. Randomized controlled trials show that the addition of cetuximab to folinic acid, 5-flourouracil, irinotecan (FOLFIRI), folinic acid, 5-flourouracil, oxaliplatin (FOLFOX) and capecitabin + oxaliplatin (CAPOX) regimens, as the first-line treatment for metastatic colorectal cancer (CRC), increases the overall survival (OS) and progression-free survival (PFS) compared to FOLFIRI, FOLFOX and CAPOX regimens alone. The aim of this study was to analyze the cost-effectiveness of different treatment programs for managing metastatic CRC with and without cetuximab in the first-line treatment of unresectable metastatic CRC in Iran. METHODS A systematic search of the literature was performed in PubMed, Centre for Reviews and Dissemination Databases and Cochrane Library to assess the effectiveness of the drug in the context of PFS, OS and the adverse events. The incremental cost-effectiveness ratio of each treatment program was calculated. An extensive sensitivity analysis was conducted on the results regarding the effectiveness. RESULTS The addition of cetuximab to FOLFIRI, FOLFOX and CAPOX programs increased PFS by 0.1, 0.042 and 0.042 years, respectively. Similarly, the addition of cetuximab to FOLFIRI, FOLFOX and CAPOX increased OS by 0.325, 0.442 and 0.442 years and also cost $212825, $202484 and $204198 individually. Whereas, based on the World Health Organisation (WHO) suggested threshold for cost-effectiveness analysis, even FOLFOX + cetuximab was very higher than the threshold in Iran (37.4 times higher). CONCLUSIONS The FOLFOX regimen + cetuximab provides lower costs per additional life years gained (more cost-effective) compared with its alternatives in the treatment of patients with unresectable metastatic CRC. However, according to the WHO indicator, none of the cetuximab regimens could be considered as cost effective for the Iranian health care market.
Collapse
Affiliation(s)
- Majid Davari
- Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Ashrafi
- Department of Hematology and Medical Oncology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammadreza Maracy
- Department of Statistic and Epidemiology, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abolfazl Aslani
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammadreza Tabatabaei
- Department of Clinical Pharmacy and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
34
|
Miyoshi Y, Noguchi K, Yanagisawa M, Taguri M, Morita S, Ikeda I, Fujinami K, Miura T, Kobayashi K, Uemura H. Nomogram for overall survival of Japanese patients with bone-metastatic prostate cancer. BMC Cancer 2015; 15:338. [PMID: 25929438 PMCID: PMC4423138 DOI: 10.1186/s12885-015-1330-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 04/21/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND We analyzed the relationship between prostate cancer outcomes and pretreatment clinical factors and developed a prognostic nomogram of overall survival (OS) of patients with bone metastasis. METHODS From 1993 to 2011, 463 consecutive patients were treated for bone-metastatic prostate cancer. Data sets from 361 patients were used to develop a nomogram (training data), and data sets of 102 patients were used for validation of the nomogram (validation data). Using the external validation data set, the nomogram was assessed for discriminatory ability, and the predictions were assessed for calibration accuracy by plotting actual survival against predicted risk. RESULTS Of the 361 patients in the training data set, 205 (56.8%) patients died, 169 (46.8%) deaths of which were due to prostate cancer. The median follow-up period was 55.2 months. In the multivariate analysis, patient age, serum prostate-specific antigen level, clinical T stage, extent of disease on bone scan, and biopsy Gleason sum were independent prognostic factors. We developed a prognostic model comprising these five factors for patients with bone-metastatic prostate cancer. This nomogram can be used to estimate 1-, 3-, and 5-year survival probability. External validation of this model using 102 validation data sets showed reasonable accuracy (concordance index, 0.719). CONCLUSION Our pretreatment prognostic nomogram might be useful for Japanese patients with bone-metastatic prostate cancer.
Collapse
Affiliation(s)
- Yasuhide Miyoshi
- Department of Urology, Yokohama City University Medical Center, Yokohama, Japan.
| | - Kazumi Noguchi
- Department of Urology, Yokohama City University Medical Center, Yokohama, Japan.
| | - Masahiro Yanagisawa
- Department of Urology, Yokohama City University School of Medicine, Yokohama, Japan.
| | - Masataka Taguri
- Department of Biostatistics and Epidemiology, Yokohama City University School of Medicine, Yokohama, Japan.
| | - Satoshi Morita
- Department of Biostatistics and Epidemiology, Yokohama City University School of Medicine, Yokohama, Japan.
| | - Ichiro Ikeda
- Department of Urology, Yokohama Minami Kyosai Hospital, Yokohama, Japan.
| | - Kiyoshi Fujinami
- Department of Urology, Yokohama City University Medical Center, Yokohama, Japan.
| | - Takeshi Miura
- Department of Urology, Kanagawa Cancer Center, Yokohama, Japan.
| | - Kazuki Kobayashi
- Department of Urology, Yokosuka Kyosai Hospital, Yokosuka, Japan.
| | - Hiroji Uemura
- Department of Urology, Yokohama City University School of Medicine, Yokohama, Japan.
| |
Collapse
|
35
|
Aziz A, Kempkensteffen C, May M, Lebentrau S, Burger M, Chun FKH, Brookman-May S. Prognostic, predictive and potential surrogate markers in castration-resistant prostate cancer. Expert Rev Anticancer Ther 2015; 15:649-66. [DOI: 10.1586/14737140.2015.1038247] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
36
|
Potthoff RF, Halabi S. A novel test to compare two treatments based on endpoints involving both nonfatal and fatal events. Pharm Stat 2015; 14:273-83. [PMID: 25894200 DOI: 10.1002/pst.1683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 03/23/2015] [Accepted: 03/23/2015] [Indexed: 11/09/2022]
Abstract
In a clinical trial comparing two treatment groups, one commonly-used endpoint is time to death. Another is time until the first nonfatal event (if there is one) or until death (if not). Both endpoints have drawbacks. The wrong choice may adversely affect the value of the study by impairing power if deaths are too few (with the first endpoint) or by lessening the role of mortality if not (with the second endpoint). We propose a compromise that provides a simple test based on the time to death if the patient has died or time since randomization augmented by an increment otherwise. The test applies the ordinary two-sample Wilcoxon statistic to these values. The formula for the increment (the same for experimental and control patients) must be specified before the trial starts. In the simplest (and perhaps most useful) case, the increment assumes only two values, according to whether or not the (surviving) patient had a nonfatal event. More generally, the increment depends on the time of the first nonfatal event, if any, and the time since randomization. The test has correct Type I error even though it does not handle censoring in a customary way. For conditions where investigators would face no easy (advance) choice between the two older tests, simulation results favor the new test. An example using a renal-cancer trial is presented.
Collapse
Affiliation(s)
- Richard F Potthoff
- Cancer Statistical Center, Duke University Medical Center, 2424 Erwin Road, Suite 802, NC, 27705, Durham, USA
| | - Susan Halabi
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, 2424 Erwin Road, Suite 802, NC, 27705, Durham, USA
| |
Collapse
|
37
|
Armstrong AJ, Halabi S. Making progress on progression in metastatic prostate cancer. J Clin Oncol 2015; 33:1322-4. [PMID: 25667271 DOI: 10.1200/jco.2014.59.4283] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
| | - Susan Halabi
- Alliance Statistics and Data Center, Duke University, Durham, NC
| |
Collapse
|
38
|
Morris MJ, Molina A, Small EJ, de Bono JS, Logothetis CJ, Fizazi K, de Souza P, Kantoff PW, Higano CS, Li J, Kheoh T, Larson SM, Matheny SL, Naini V, Burzykowski T, Griffin TW, Scher HI, Ryan CJ. Radiographic progression-free survival as a response biomarker in metastatic castration-resistant prostate cancer: COU-AA-302 results. J Clin Oncol 2015; 33:1356-63. [PMID: 25624432 DOI: 10.1200/jco.2014.55.3875] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Progression-free survival (PFS) in metastatic castration-resistant prostate cancer (mCRPC) trials has been inconsistently defined and poorly associated with overall survival (OS). A reproducible quantitative definition of radiographic PFS (rPFS) was tested for association with a coprimary end point of OS in a randomized trial of abiraterone in patients with mCRPC. PATIENTS AND METHODS rPFS was defined as ≥ two new lesions on an 8-week bone scan plus two additional lesions on a confirmatory scan, ≥ two new confirmed lesions on any scan ≥ 12 weeks after random assignment, and/or progression in nodes or viscera on cross-sectional imaging, or death. rPFS was assessed by independent review at 15% of deaths and by investigator review at 15% and 40% of deaths. rPFS and OS association was evaluated by Spearman's correlation. RESULTS A total of 1,088 patients were randomly assigned to abiraterone plus prednisone or prednisone alone. At first interim analysis, the hazard ratio (HR) by independent review was 0.43 (95% CI, 0.35 to 0.52; P < .001; abiraterone plus prednisone: median rPFS, not estimable; prednisone: median rPFS, 8.3 months). Similar HRs were obtained by investigator review at the first two interim analyses (HR, 0.49; 95% CI, 0.41 to 0.60; P < .001 and HR, 0.53; 95% CI, 0.45 to 0.62; P < .001, respectively), validating the imaging data assay used. Spearman's correlation coefficient between rPFS and OS was 0.72. CONCLUSION rPFS was highly consistent and highly associated with OS, providing initial prospective evidence on further developing rPFS as an intermediate end point in mCRPC trials.
Collapse
Affiliation(s)
- Michael J Morris
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium.
| | - Arturo Molina
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Eric J Small
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Johann S de Bono
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Christopher J Logothetis
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Karim Fizazi
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Paul de Souza
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Philip W Kantoff
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Celestia S Higano
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Jinhui Li
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Thian Kheoh
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Steven M Larson
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Shannon L Matheny
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Vahid Naini
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Tomasz Burzykowski
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Thomas W Griffin
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Howard I Scher
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Charles J Ryan
- Michael J. Morris, Steven M. Larson, and Howard I. Scher, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; Arturo Molina, Thian Kheoh, Shannon L. Matheny, Vahid Naini, and Thomas W. Griffin, Janssen Research & Development, Los Angeles; Eric J. Small and Charles J. Ryan, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; Johann S. de Bono, Institute for Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom; Christopher J. Logothetis, MD Anderson Cancer Center, Houston, TX; Karim Fizazi, Institut Gustave Roussy, University of Paris Sud, Villejuif, France; Paul de Souza, University of Western Sydney School of Medicine, Ingham Institute, Liverpool, New South Wales, Australia; Philip W. Kantoff, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Celestia S. Higano, University of Washington, Seattle, WA; Jinhui Li, Janssen Research & Development, Raritan, NJ; and Tomasz Burzykowski, International Drug Development Institute, Louvain-la-Neuve, Belgium
| |
Collapse
|
39
|
Bria E, Massari F, Maines F, Pilotto S, Bonomi M, Porta C, Bracarda S, Heng D, Santini D, Sperduti I, Giannarelli D, Cognetti F, Tortora G, Milella M. Progression-free survival as primary endpoint in randomized clinical trials of targeted agents for advanced renal cell carcinoma. Correlation with overall survival, benchmarking and power analysis. Crit Rev Oncol Hematol 2015; 93:50-9. [DOI: 10.1016/j.critrevonc.2014.08.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 06/08/2014] [Accepted: 08/08/2014] [Indexed: 10/24/2022] Open
|
40
|
Abstract
Angiogenesis is a very complex physiological process, which involves multiple pathways that are dependent on the homeostatic balance between the growth factors (stimulators and inhibitors). This tightly controlled process is stimulated by angiogenic factors, which are present within the tumor and surrounding tumor-associated stromal cells. The dependence of tumor propagation, invasion and metastasis on angiogenesis makes the inhibitors of new blood vessel formation attractive drugs for treating the malignancies. Angiogenesis can be disrupted by several distinct mechanisms: by inhibiting endothelial cells, by interrupting the signaling pathways or by inhibiting other activators of angiogenesis. This strategy has shown therapeutic benefit in several types of solid tumors, leading to Food and Drug Administration (FDA) approval of anti-angiogenic agents in the treatment of kidney, non-small cell lung, colon and brain cancers. Although no angiogenesis inhibitors have been approved for patients with metastatic prostate cancer, therapies that target new blood vessel formation are still an emerging and promising area of prostate cancer research.
Collapse
Affiliation(s)
| | - Yu-Ning Wong
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
41
|
Matulonis UA, Oza AM, Ho TW, Ledermann JA. Intermediate clinical endpoints: A bridge between progression-free survival and overall survival in ovarian cancer trials. Cancer 2014; 121:1737-46. [DOI: 10.1002/cncr.29082] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 07/08/2014] [Accepted: 07/15/2014] [Indexed: 11/07/2022]
Affiliation(s)
| | - Amit M. Oza
- Princess Margaret Cancer Centre; Toronto Canada
| | | | | |
Collapse
|
42
|
Kim W, Ryan CJ. Quo vadis: advanced prostate cancer-clinical care and clinical research in the era of multiple androgen receptor-directed therapies. Cancer 2014; 121:361-71. [PMID: 25236176 DOI: 10.1002/cncr.28929] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 06/20/2014] [Accepted: 06/30/2014] [Indexed: 11/07/2022]
Abstract
The novel androgen receptor-directed therapies abiraterone acetate and enzalutamide, having demonstrated improved survival in randomized phase 3 studies of men with metastatic castration-resistant prostate cancer, have ushered in a new era in the treatment of this disease. Additional novel androgen receptor-directed therapies, such as ARN-509 and orteronel, are in various phases of clinical trials and development. However, the emergence of therapeutic resistance and clinical disease progression is inevitable. Although advances in genomic technologies have led to unprecedented understanding of the biology of castration-resistant prostate cancer, efforts only now are underway to elucidate the mechanisms of resistance associated with abiraterone and enzalutamide. A tremendous challenge in the near future will be to determine the optimal sequence or combination of therapies to overcome resistance mechanisms. In this review, the current landscape of androgen receptor-directed therapies and future directions necessary to enhance their clinical efficacy for the maximal benefit of patients are discussed.
Collapse
Affiliation(s)
- Won Kim
- Helen Diller Family Comprehensive Cancer Center, University of California-San Francisco, San Francisco, California
| | | |
Collapse
|
43
|
Cetnar JP, Beer TM. Personalizing prostate cancer therapy: the way forward. Drug Discov Today 2014; 19:1483-7. [PMID: 25046532 DOI: 10.1016/j.drudis.2014.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 07/11/2014] [Indexed: 01/12/2023]
Abstract
Advances in genomic sequencing and molecular characterization are improving our understanding of the biology of prostate cancer and challenging us to translate emerging data into meaningful clinical outcomes. Several recently approved treatments for advanced prostate cancer extend survival; however, these therapies are not personalized based on predictive biomarkers. Innovative strategies for early phase drug testing that harness our growing knowledge of important prognostic markers and emerging predictive biomarkers are needed. In this review we discuss new strategies to assess drug response in early phase clinical trial testing.
Collapse
Affiliation(s)
- Jeremy P Cetnar
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR, USA
| | - Tomasz M Beer
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR, USA.
| |
Collapse
|
44
|
Sonpavde G, Pond GR, Armstrong AJ, Galsky MD, Leopold L, Wood BA, Wang SL, Paolini J, Chen I, Chow-Maneval E, Mooney DJ, Lechuga M, Smith MR, Michaelson MD. Radiographic progression by Prostate Cancer Working Group (PCWG)-2 criteria as an intermediate endpoint for drug development in metastatic castration-resistant prostate cancer. BJU Int 2014; 114:E25-E31. [DOI: 10.1111/bju.12589] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Guru Sonpavde
- Department of Medicine; Division of Hematology-Oncology; University of Alabama; Birmingham (UAB) Comprehensive Cancer Center; Birmingham AL USA
| | - Gregory R. Pond
- Department of Biostatistics; McMaster University; Hamilton ON Canada
| | - Andrew J. Armstrong
- Department of Medicine; Division of Hematology-Oncology; Duke Prostate Center; Durham NC USA
| | - Matthew D. Galsky
- Department of Medicine; Division of Hematology-Oncology; Mt Sinai Tisch Cancer Institute; New York NY USA
| | | | | | - Shaw-Ling Wang
- Pfizer Inc.; Madison NJ USA
- ICON Clinical Research Inc.; San Diego CA USA
| | | | - Isan Chen
- Pfizer Inc.; Madison NJ USA
- Seragon Pharmaceuticals; San Diego CA USA
| | - Edna Chow-Maneval
- Pfizer Inc.; Madison NJ USA
- Seragon Pharmaceuticals; San Diego CA USA
| | - David J. Mooney
- Department of Medicine; Division of Hematology-Oncology; University of Alabama; Birmingham (UAB) Comprehensive Cancer Center; Birmingham AL USA
| | | | - Matthew R. Smith
- Department of Medicine; Division of Hematology-Oncology; Massachusetts General Hospital and the Harvard Medical School; Boston MA USA
| | - M. Dror Michaelson
- Department of Medicine; Division of Hematology-Oncology; Massachusetts General Hospital and the Harvard Medical School; Boston MA USA
| |
Collapse
|
45
|
Algarra R, Hevia M, Tienza A, Merino I, Velis JM, Zudaire J, Robles JE, Pascual I. Survival analysis of patients with biochemical relapse after radical prostatectomy treated with androgen deprivation: Castration-resistance influential factors. Can Urol Assoc J 2014; 8:E333-41. [PMID: 24940460 DOI: 10.5489/cuaj.1665] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
INTRODUCTION We evaluate the prognosis of patients with biochemical recurrence (BCR) treated with androgen deprivation therapy (ADT) and to determine the influential factors to castration resistance (CR) and death. METHODS From a series of 1310 patients with T1-T2 prostate cancer treated with radical prostatectomy between 1989 and 2012, 371 had BCR. Patients with lymph node involvement were excluded. We analyzed only the 159 treated with salvage ADT. At the end of the study, 77 (48%) had developed CR. RESULTS The median follow-up to CR was 9.2 years. The CR-resistant free survival (RFS) was 76 ± 3%, 62 ± 3% and 43 ± 9% in 5, 10 and 15 years, respectively. The RFS median time was 14 years. In the multivariate study, the prostate-specific antigen (PSA) doubling time (PSA-DT) was <6 months (p = 0.01) (hazard ratio [HR] 3; 95% confidence interval [CI] 1.4-6.8, p = 0.007); seminal vesicle involvement (HR 3.1; 95% CI 1.5-6.2, p = 0.01) and PSA velocity in ng/mL/year (HR 1.3; 95% CI 1.1-1.5, p = 0.002) with better cut-off points of 0.84 ng/mL/year (p = 0.04) (HR 4; 95% CI 1.7-9.4, p = 0.001) were influential variables. Specific survival (SS) at 5, 10 and 15 years since surgery was 96 ± 1, 85 ± 2 and 76 ± 4, respectively. The time of CR to death was 30 ± 6% at 5 years, with the median at 3.2 years. In the multivariate only Ki 67 (HR 1.04; 95% CI 1.005-1.08, p = 0.02) had an independent influence. CONCLUSIONS In BCR patients treated with ADT, the median to CR was 14 years. PSA-DT <6 months, PSA velocity (ng/mL/year) and seminal vesicle involvement were influential variables. From the CR, the median time to death was 3.2 years. Ki-67 marker was an independent influence.
Collapse
Affiliation(s)
- Rubén Algarra
- Department of Urology, University of Navarra Clinic, Navarra, Spain
| | - Mateo Hevia
- Department of Urology, University of Navarra Clinic, Navarra, Spain
| | - Antonio Tienza
- Department of Urology, University of Navarra Clinic, Navarra, Spain
| | - Imanol Merino
- Department of Urology, University of Navarra Clinic, Navarra, Spain
| | - José María Velis
- Department of Urology, University of Navarra Clinic, Navarra, Spain
| | - Javier Zudaire
- Department of Urology, University of Navarra Clinic, Navarra, Spain
| | | | - Ignacio Pascual
- Department of Urology, University of Navarra Clinic, Navarra, Spain
| |
Collapse
|
46
|
Lee D, Porter J, Gladwell D, Brereton N, Nielsen SK. A cost-utility analysis of degarelix in the treatment of advanced hormone-dependent prostate cancer in the United Kingdom. J Med Econ 2014; 17:233-47. [PMID: 24568188 DOI: 10.3111/13696998.2014.893240] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To determine the cost-effectiveness of the treatment of advanced hormone-dependent prostate cancer with degarelix compared to luteinizing hormone-releasing hormone (LHRH) agonists in the UK using the latest available evidence and the model submitted to AWMSG. METHODS A cost-effectiveness model was developed from the perspective of the UK National Health Service evaluating monthly injection of degarelix against 3-monthly leuprorelin therapy plus anti-androgen flare cover for the first-line treatment of patients with advanced (locally advanced or metastatic) hormone-dependent prostate cancer. A Markov process model was constructed using the patient population characteristics and efficacy information from the CS21 Phase III clinical trial and associated extension study (CS21A). The intention-to-treat (ITT) population and a high-risk sub-group with a PSA level >20 ng/mL were modeled. RESULTS In the base-case analysis using the patient access scheme (PAS) price, degarelix was dominant compared to leuprorelin with cost savings of £3633 in the ITT population and £4310 in the PSA > 20 ng/mL sub-group. The chance of being cost-effective was 95% in the ITT population and 96% in the PSA > 20 ng/mL sub-group at a threshold of £20,000 per quality-adjusted life-year (QALY). In addition, degarelix remained dominant when PSA progression was assumed equal and only the benefits of preventing testosterone flare were taken into account. Treatment with degarelix also remained dominant in both populations when the list price was used. The additional investment required to treat patients with degarelix could be offset in 19 months for the ITT population and 13 months for the PSA > 20 ng/mL population. The model was most sensitive to the hazard ratio assumed for PSA progression between degarelix and leuprorelin and the quality-of-life (utility) of patients receiving palliative care. CONCLUSION Degarelix is likely to be cost-effective compared to leuprorelin plus anti-androgen flare cover in the first-line treatment of advanced hormone-dependent prostate cancer.
Collapse
|
47
|
Trendel JA. The hurdle of antiandrogen drug resistance: drug design strategies. Expert Opin Drug Discov 2013; 8:1491-501. [PMID: 24206221 DOI: 10.1517/17460441.2013.855194] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Prostate cancer is the second most common cancer death in men after lung cancer, due to distant metastases. While distant prostate cancer is typically castrate resistant, it is not necessarily androgen independent. For this reason, a review of the literature regarding the pathways involved in androgen signaling and therapeutic regimens to treat distant metastases is beneficial to increasing the survival rate of prostate cancer patients. AREAS COVERED In this article, the author reviews the literature from the past decade covering metastatic hormone refractory prostate cancer with the aim to examine and identify pathways, therapeutic targets and current therapies for treating castrate-resistant disease. As this area is lacking, the author aims to provide the reader with knowledge of the molecular consequences of castrate resistant prostate cancer, the current treatment paradigms and future directions. EXPERT OPINION While there have been advances in the treatment of castrate resistant prostate cancer, only minimal advances have been made in overall survival rate. Due to aberrant mutations and activation in the androgen receptor gene, and the complexity of cell signaling within prostate cancer, the androgen receptor should remain a main target for drug discovery efforts. This author believes that designing compounds that will reduce the activation of the androgen receptor may hold the key to a cure in the future.
Collapse
Affiliation(s)
- Jill A Trendel
- University of Toledo, Center for Drug Design and Development , 3000 Arlington Ave MS 1015 Toledo, OH 43614 , USA +1 419 383 1536 ;
| |
Collapse
|
48
|
Gallardo E, Arranz JÁ, Maroto JP, León LÁ, Bellmunt J. Expert opinion on chemotherapy use in castration-resistant prostate cancer progressing after docetaxel. Crit Rev Oncol Hematol 2013; 88:357-67. [PMID: 23867574 DOI: 10.1016/j.critrevonc.2013.06.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 05/28/2013] [Accepted: 06/19/2013] [Indexed: 01/08/2023] Open
Abstract
The term castration-resistant prostate cancer (CRPC) encompasses a wide variety of patients with different prognoses. The combination of docetaxel and prednisone is considered as the standard first-line chemotherapy. For years, patients progressing on docetaxel have been managed with second- and third-line hormone therapies, re-treatment with docetaxel, or combined mitoxantrone and prednisone. Recently published results of four studies using different drugs: cabazitaxel (CBZ), abiraterone (AA), enzalutamide (ENZ), and radium 223, showed an increased survival in such patients. In this article, authors make some considerations about criteria guiding the choice of a second-line chemotherapy after docetaxel in patients with metastatic CRPC, and propose an algorithm based on scientific evidence and consensus for rational use of cabazitaxel in this scenario.
Collapse
Affiliation(s)
- Enrique Gallardo
- Department of Oncology, Parc Taulí Sabadell Hospital Universitari, Sabadell, Barcelona, Spain.
| | | | | | | | | |
Collapse
|
49
|
Halabi S, Rini B, Escudier B, Stadler WM, Small EJ. Progression-free survival as a surrogate endpoint of overall survival in patients with metastatic renal cell carcinoma. Cancer 2013; 120:52-60. [PMID: 24347384 DOI: 10.1002/cncr.28221] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 05/16/2013] [Accepted: 05/21/2013] [Indexed: 11/05/2022]
Abstract
BACKGROUND The current study was conducted to investigate the dependence between progression-free survival (PFS) and overall survival (OS) in patients with metastatic renal cell carcinoma (mRCC) and to explore whether PFS can be used as an intermediate endpoint of OS in this patient population. METHODS A total of 1381 patients from 2 prospective phase 3 trials (Cancer and Leukemia Group B [CALGB] 90206 and AVOREN) of interferon-alpha with or without bevacizumab were analyzed. Both trials recruited previously untreated patients with clear cell mRCC with an Eastern Cooperative Oncology Group performance status of 0 to 2; adequate bone marrow, hepatic, cardiac, and renal function; and controlled blood pressure. The CALGB study served as the training data set, and the AVOREN study served as the testing data set. The dependence between PFS and OS was investigated using the Kendall tau for bivariate time-to-event endpoints. RESULTS In the training data set, the median OS times among patients who experienced progressive disease at 3 months or 6 months were 6 months and 8 months, respectively, compared with 25 months and 30 months, respectively, (P < .001) in patients who did not develop disease progress. The adjusted hazard ratios (HR) were 2.6 (P < .0001) and 2.8 (P < .0001), respectively, for patients who did and did not progress at 3 months or 6 months. The dependence between PFS and OS was 0.53. These associations were confirmed in the testing data set. CONCLUSIONS In patients with mRCC who were treated with interferon-alpha with or without bevacizumab, the PFS at 3 months and 6 months was found to be predictive of OS. A high dependence between PFS and OS was observed, suggesting that PFS may be used as a surrogate endpoint for OS. Although this is a novel observation for RCC, these findings require validation in patients with mRCC who are treated with other targeted agents.
Collapse
Affiliation(s)
- Susan Halabi
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina; Alliance Statistical and Data Center, Duke University, Durham, North Carolina
| | | | | | | | | |
Collapse
|
50
|
Becker A, Eichelberg C, Sun M. Progression-free survival: Does a correlation with survival justify its role as a surrogate clinical endpoint? Cancer 2013; 120:7-10. [DOI: 10.1002/cncr.28378] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2013] [Revised: 08/25/2013] [Accepted: 09/03/2013] [Indexed: 11/07/2022]
Affiliation(s)
- Andreas Becker
- Department of Urology; University Hospital Hamburg-Eppendorf; Hamburg Germany
- Martini-Clinic; Prostate Cancer Center; Hamburg Germany
| | | | - Maxine Sun
- Cancer Prognostics and Health Outcomes Unit, Department of Urology; University of Montreal Health Centre; Montreal Quebec Canada
| |
Collapse
|