151
|
Colliaux J, Kharchi L, Vincendeau S, Simon A, Perdrieux M, Le Prisé É, Bellisant É, Castelli J, de Crevoisier R. [Expected benefit of lymph node and seminal vesical dissection to decrease high-risk prostate cancer radiotherapy]. Cancer Radiother 2016; 20:347-56. [PMID: 27344535 DOI: 10.1016/j.canrad.2016.04.006] [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: 11/30/2015] [Revised: 04/07/2016] [Accepted: 04/15/2016] [Indexed: 11/28/2022]
Abstract
PURPOSE In case of pelvic lymph node and seminal vesicle dissection followed by prostate cancer intensity-modulated radiotherapy, the objective of the study was to evaluate the dosimetric benefit of reducing the target volume. PATIENTS AND METHODS A total of 25 patients with high-risk prostate cancer had surgery first followed by intensity-modulated radiotherapy and androgen deprivation. Four treatment planning were simulated for each patient, based on two CT scans performed before and after surgery. The target volumes were: prostate-seminal vesicles-lymph nodes, prostate-lymph nodes, prostate-seminal vesicles and prostate only. The total dose was 46Gy in the seminal vesicles and lymph nodes, and 80Gy in the prostate. RESULTS Compared to prostate target volume only, the addition of seminal vesicles and lymph nodes multiplied by a factor of 1.6 and 6.5 the target volume, respectively. Decreasing the target volume from prostate-seminal vesicles-lymph nodes to prostate-seminal vesicles, to prostate only decreased the rectal wall mean dose from 49Gy to 42Gy, to 36Gy, and the risk of late rectal bleeding from 4.4% to 3.2%, to 2.4% (P<0.05), respectively. The bladder wall mean dose decreased from 51Gy to 40Gy, to 35Gy (P<0,05), respectively. Not irradiating the lymph nodes decreased the absolute risk of diarrhea by 11%. CONCLUSION Lymph node and seminal vesicle dissection before prostate cancer intensity-modulated radiotherapy allows decreasing moderately the risk of digestive toxicity.
Collapse
Affiliation(s)
- J Colliaux
- Département de radiothérapie, centre régional de lutte contre le cancer Eugène-Marquis, avenue de la Bataille-Flandres-Dunkerque, 35000 Rennes, France; Université de Rennes 1, centre investigation clinique, CHU Pontchaillou, 2, rue Henri-Le Guilloux, 35000 Rennes, France
| | - L Kharchi
- Département de radiothérapie, centre régional de lutte contre le cancer Eugène-Marquis, avenue de la Bataille-Flandres-Dunkerque, 35000 Rennes, France; Inserm, U1099, laboratoire de traitement du signal et de l'image, 263, avenue du Général-Leclerc, 35042 Rennes, France
| | - S Vincendeau
- Service d'urologie, CHU Pontchaillou, 2, rue Henri-Le-Guilloux, 35000 Rennes, France
| | - A Simon
- Inserm, U1099, laboratoire de traitement du signal et de l'image, 263, avenue du Général-Leclerc, 35042 Rennes, France
| | - M Perdrieux
- Département de radiothérapie, centre régional de lutte contre le cancer Eugène-Marquis, avenue de la Bataille-Flandres-Dunkerque, 35000 Rennes, France
| | - É Le Prisé
- Département de radiothérapie, centre régional de lutte contre le cancer Eugène-Marquis, avenue de la Bataille-Flandres-Dunkerque, 35000 Rennes, France
| | - É Bellisant
- Université de Rennes 1, centre investigation clinique, CHU Pontchaillou, 2, rue Henri-Le Guilloux, 35000 Rennes, France
| | - J Castelli
- Département de radiothérapie, centre régional de lutte contre le cancer Eugène-Marquis, avenue de la Bataille-Flandres-Dunkerque, 35000 Rennes, France; Université de Rennes 1, centre investigation clinique, CHU Pontchaillou, 2, rue Henri-Le Guilloux, 35000 Rennes, France
| | - R de Crevoisier
- Département de radiothérapie, centre régional de lutte contre le cancer Eugène-Marquis, avenue de la Bataille-Flandres-Dunkerque, 35000 Rennes, France; Inserm, U1099, laboratoire de traitement du signal et de l'image, 263, avenue du Général-Leclerc, 35042 Rennes, France; Université de Rennes 1, centre investigation clinique, CHU Pontchaillou, 2, rue Henri-Le Guilloux, 35000 Rennes, France.
| |
Collapse
|
152
|
Juloori A, Shah C, Stephans K, Vassil A, Tendulkar R. Evolving Paradigm of Radiotherapy for High-Risk Prostate Cancer: Current Consensus and Continuing Controversies. Prostate Cancer 2016; 2016:2420786. [PMID: 27313896 PMCID: PMC4893567 DOI: 10.1155/2016/2420786] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 04/16/2016] [Accepted: 04/24/2016] [Indexed: 11/17/2022] Open
Abstract
High-risk prostate cancer is an aggressive form of the disease with an increased risk of distant metastasis and subsequent mortality. Multiple randomized trials have established that the combination of radiation therapy and long-term androgen deprivation therapy improves overall survival compared to either treatment alone. Standard of care for men with high-risk prostate cancer in the modern setting is dose-escalated radiotherapy along with 2-3 years of androgen deprivation therapy (ADT). There are research efforts directed towards assessing the efficacy of shorter ADT duration. Current research has been focused on assessing hypofractionated and stereotactic body radiation therapy (SBRT) techniques. Ongoing randomized trials will help assess the utility of pelvic lymph node irradiation. Research is also focused on multimodality therapy with addition of a brachytherapy boost to external beam radiation to help improve outcomes in men with high-risk prostate cancer.
Collapse
Affiliation(s)
- Aditya Juloori
- Cleveland Clinic, Taussig Cancer Institute, Department of Radiation Oncology, Cleveland, OH, USA
| | - Chirag Shah
- Cleveland Clinic, Taussig Cancer Institute, Department of Radiation Oncology, Cleveland, OH, USA
| | - Kevin Stephans
- Cleveland Clinic, Taussig Cancer Institute, Department of Radiation Oncology, Cleveland, OH, USA
| | - Andrew Vassil
- Cleveland Clinic, Taussig Cancer Institute, Department of Radiation Oncology, Strongsville, OH, USA
| | - Rahul Tendulkar
- Cleveland Clinic, Taussig Cancer Institute, Department of Radiation Oncology, Cleveland, OH, USA
| |
Collapse
|
153
|
Mercado C, Kress MA, Cyr RA, Chen LN, Yung TM, Bullock EG, Lei S, Collins BT, Satinsky AN, Harter KW, Suy S, Dritschilo A, Lynch JH, Collins SP. Intensity-Modulated Radiation Therapy with Stereotactic Body Radiation Therapy Boost for Unfavorable Prostate Cancer: The Georgetown University Experience. Front Oncol 2016; 6:114. [PMID: 27200300 PMCID: PMC4858516 DOI: 10.3389/fonc.2016.00114] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 04/20/2016] [Indexed: 12/29/2022] Open
Abstract
Purpose/objective(s) Stereotactic body radiation therapy (SBRT) is emerging as a minimally invasive alternative to brachytherapy to deliver highly conformal, dose-escalated radiation therapy (RT) to the prostate. SBRT alone may not adequately cover the tumor extensions outside the prostate commonly seen in unfavorable prostate cancer. External beam radiation therapy (EBRT) with high dose rate brachytherapy boost is a proven effective therapy for unfavorable prostate cancer. This study reports on early prostate-specific antigen and prostate cancer-specific quality of life (QOL) outcomes in a cohort of unfavorable patients treated with intensity-modulated radiation therapy (IMRT) and SBRT boost. Materials/methods Prostate cancer patients treated with SBRT (19.5 Gy in three fractions) followed by fiducial-guided IMRT (45–50.4 Gy) from March 2008 to September 2012 were included in this retrospective review of prospectively collected data. Biochemical failure was assessed using the Phoenix definition. Patients completed the expanded prostate cancer index composite (EPIC)-26 at baseline, 1 month after the completion of RT, every 3 months for the first year, then every 6 months for a minimum of 2 years. Results One hundred eight patients (4 low-, 45 intermediate-, and 59 high-risk) with median age of 74 years completed treatment, with median follow-up of 4.4 years. Sixty-four percent of the patients received androgen deprivation therapy prior to the initiation of RT. The 3-year actuarial biochemical control rates were 100 and 89.8% for intermediate- and high-risk patients, respectively. At the initiation of RT, 9 and 5% of men felt their urinary and bowel function was a moderate to big problem, respectively. Mean EPIC urinary and bowel function and bother scores exhibited transient declines, with subsequent return to near baseline. At 2 years posttreatment, 13.7 and 5% of men felt their urinary and bowel function was a moderate to big problem, respectively. Conclusion At 3-year follow-up, biochemical control was favorable. Acute urinary and bowel symptoms were comparable to conventionally fractionated IMRT and brachytherapy. Patients recovered to near their baseline urinary and bowel function by 2 years posttreatment. A combination of IMRT with SBRT boost is well tolerated with minimal impact on prostate cancer-specific QOL.
Collapse
Affiliation(s)
- Catherine Mercado
- Department of Radiation Medicine, Georgetown University Hospital , Washington, DC , USA
| | - Marie-Adele Kress
- Department of Radiation Medicine, Georgetown University Hospital , Washington, DC , USA
| | - Robyn A Cyr
- Department of Radiation Medicine, Georgetown University Hospital , Washington, DC , USA
| | - Leonard N Chen
- Department of Radiation Medicine, Georgetown University Hospital , Washington, DC , USA
| | - Thomas M Yung
- Department of Radiation Medicine, Georgetown University Hospital , Washington, DC , USA
| | - Elizabeth G Bullock
- Department of Radiation Medicine, Georgetown University Hospital , Washington, DC , USA
| | - Siyuan Lei
- Department of Radiation Medicine, Georgetown University Hospital , Washington, DC , USA
| | - Brian T Collins
- Department of Radiation Medicine, Georgetown University Hospital , Washington, DC , USA
| | - Andrew N Satinsky
- Department of Radiation Medicine, Georgetown University Hospital , Washington, DC , USA
| | - K William Harter
- Department of Radiation Medicine, Georgetown University Hospital , Washington, DC , USA
| | - Simeng Suy
- Department of Radiation Medicine, Georgetown University Hospital , Washington, DC , USA
| | - Anatoly Dritschilo
- Department of Radiation Medicine, Georgetown University Hospital , Washington, DC , USA
| | - John H Lynch
- Department of Urology, Georgetown University Hospital , Washington, DC , USA
| | - Sean P Collins
- Department of Radiation Medicine, Georgetown University Hospital , Washington, DC , USA
| |
Collapse
|
154
|
Jereczek-Fossa BA, Ciardo D, Ferrario S, Fossati P, Fanetti G, Zerini D, Zannoni D, Fodor C, Gerardi MA, Surgo A, Muto M, Cambria R, De Cobelli O, Orecchia R. No increase in toxicity of pelvic irradiation when intensity modulation is employed: clinical and dosimetric data of 208 patients treated with post-prostatectomy radiotherapy. Br J Radiol 2016; 89:20150985. [PMID: 27109736 DOI: 10.1259/bjr.20150985] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To compare the toxicity of image-guided intensity-modulated radiotherapy (IG-IMRT) to the pelvis or prostate bed (PB) only. To test the hypothesis that the potentially injurious effect of pelvic irradiation can be counterbalanced by reduced irradiated normal tissue volume using IG-IMRT. METHODS Between February 2010 and February 2012, 208 patients with prostate cancer were treated with adjuvant or salvage IG-IMRT to the PB (102 patients, Group PB) or the pelvis and prostate bed (P) (106 patients, Group P). The Radiation Therapy Oncology Group/European Organization for Research and Treatment of Cancer criteria were used to evaluate toxicity. RESULTS Median follow-up was 27 months. Toxicity G ≥ 2 in Group PB: in the bowel acute and late toxicities were 11.8% and 10%, respectively; urinary acute and late toxicities were 10.8% and 15%, respectively. Toxicity G ≥ 2 in Group P: in the bowel acute and late toxicities were both 13.2%; urinary acute and late toxicities were 13.2% and 15.1%, respectively. No statistical difference in acute or late toxicity between the groups was found (bowel: p = 0.23 and p = 0.89 for acute and late toxicity, respectively; urinary: p = 0.39 and p = 0.66 for acute and late toxicity, respectively). Of the clinical variables, only previous abdominal surgery was correlated with acute bowel toxicity. Dosimetric parameters that correlated with bowel toxicity were identified. CONCLUSION The toxicity rates were low and similar in both groups, suggesting that IG-IMRT allows for a safe post-operative irradiation of larger volumes. Further investigation is warranted to exclude bias owing to non-randomized character of the study. ADVANCES IN KNOWLEDGE Our report shows that modern radiotherapy technology and careful planning allow maintaining the toxicity of pelvic lymph node treatment at the acceptable level, as it is in the case of PB radiotherapy.
Collapse
Affiliation(s)
- Barbara A Jereczek-Fossa
- 1 Division of Radiation Oncology, European Institute of Oncology, Milan, Italy.,2 Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Delia Ciardo
- 1 Division of Radiation Oncology, European Institute of Oncology, Milan, Italy
| | - Silvia Ferrario
- 1 Division of Radiation Oncology, European Institute of Oncology, Milan, Italy.,2 Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Piero Fossati
- 1 Division of Radiation Oncology, European Institute of Oncology, Milan, Italy.,2 Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Giuseppe Fanetti
- 1 Division of Radiation Oncology, European Institute of Oncology, Milan, Italy.,2 Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Dario Zerini
- 1 Division of Radiation Oncology, European Institute of Oncology, Milan, Italy
| | | | - Cristiana Fodor
- 1 Division of Radiation Oncology, European Institute of Oncology, Milan, Italy
| | - Marianna A Gerardi
- 1 Division of Radiation Oncology, European Institute of Oncology, Milan, Italy.,2 Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Alessia Surgo
- 1 Division of Radiation Oncology, European Institute of Oncology, Milan, Italy.,2 Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Matteo Muto
- 1 Division of Radiation Oncology, European Institute of Oncology, Milan, Italy.,2 Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Raffaella Cambria
- 4 Division of Medical Physics, European Institute of Oncology, Milan, Italy
| | - Ottavio De Cobelli
- 2 Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy.,5 Division of Urology, European Institute of Oncology, Milan, Italy
| | - Roberto Orecchia
- 1 Division of Radiation Oncology, European Institute of Oncology, Milan, Italy.,2 Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| |
Collapse
|
155
|
Bellefqih S, Hadadi K, Mezouri I, Maghous A, Marnouche E, Andaloussi K, Elmarjany M, Sifat H, Mansouri H, Benjaafar N. Association de radiothérapie et d’hormonothérapie dans la prise en charge des cancers localisés de la prostate : où en est-on ? Cancer Radiother 2016; 20:141-50. [DOI: 10.1016/j.canrad.2015.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/21/2015] [Accepted: 12/24/2015] [Indexed: 12/12/2022]
|
156
|
Kim YJ, Park JH, Yun IH, Kim YS. A prospective comparison of acute intestinal toxicity following whole pelvic versus small field intensity-modulated radiotherapy for prostate cancer. Onco Targets Ther 2016; 9:1319-25. [PMID: 27022287 PMCID: PMC4790507 DOI: 10.2147/ott.s96646] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose To compare the acute intestinal toxicity of whole pelvic (WP) and small field (SF) intensity-modulated radiotherapy (IMRT) for prostate cancer using dosimetric and metabolic parameters as well as clinical findings. Methods Patients who received IMRT in either a definitive or postoperative setting were prospectively enrolled. Target volume and organs at risk including intestinal cavity (IC) were delineated in every patient by a single physician. The IC volume that received a 10–50 Gy dose at 5-Gy intervals (V10–V50) and the percentage of irradiated volume as a fraction of total IC volume were calculated. Plasma citrulline levels, as an objective biological marker, were checked at three time points: baseline and after exposure to 30 Gy and 60 Gy. Results Of the 41 patients, only six experienced grade 1 acute intestinal toxicity. Although all dose–volume parameters were significantly worse following WP than SF IMRT, there was no statistically significant relationship between these dosimetric parameters and clinical symptoms. Plasma citrulline levels did not show a serial decrease by radiotherapy volume difference (WP versus SF) and were not relevant to the irradiated doses. Conclusion Given that WP had comparable acute intestinal toxicities to those associated with SF, WP IMRT appears to be a feasible approach for the treatment of prostate cancer despite dosimetric disadvantages.
Collapse
Affiliation(s)
- Yeon Joo Kim
- Department of Radiation Oncology, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Republic of Korea
| | - Jin-Hong Park
- Department of Radiation Oncology, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Republic of Korea
| | - In-Ha Yun
- Department of Radiation Oncology, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Republic of Korea
| | - Young Seok Kim
- Department of Radiation Oncology, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Republic of Korea
| |
Collapse
|
157
|
James ND, Spears MR, Clarke NW, Dearnaley DP, Mason MD, Parker CC, Ritchie AWS, Russell JM, Schiavone F, Attard G, de Bono JS, Birtle A, Engeler DS, Elliott T, Matheson D, O'Sullivan J, Pudney D, Srihari N, Wallace J, Barber J, Syndikus I, Parmar MKB, Sydes MR. Failure-Free Survival and Radiotherapy in Patients With Newly Diagnosed Nonmetastatic Prostate Cancer: Data From Patients in the Control Arm of the STAMPEDE Trial. JAMA Oncol 2016; 2:348-57. [PMID: 26606329 PMCID: PMC4789485 DOI: 10.1001/jamaoncol.2015.4350] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
IMPORTANCE The natural history of patients with newly diagnosed high-risk nonmetastatic (M0) prostate cancer receiving hormone therapy (HT) either alone or with standard-of-care radiotherapy (RT) is not well documented. Furthermore, no clinical trial has assessed the role of RT in patients with node-positive (N+) M0 disease. The STAMPEDE Trial includes such individuals, allowing an exploratory multivariate analysis of the impact of radical RT. OBJECTIVE To describe survival and the impact on failure-free survival of RT by nodal involvement in these patients. DESIGN, SETTING, AND PARTICIPANTS Cohort study using data collected for patients allocated to the control arm (standard-of-care only) of the STAMPEDE Trial between October 5, 2005, and May 1, 2014. Outcomes are presented as hazard ratios (HRs) with 95% CIs derived from adjusted Cox models; survival estimates are reported at 2 and 5 years. Participants were high-risk, hormone-naive patients with newly diagnosed M0 prostate cancer starting long-term HT for the first time. Radiotherapy is encouraged in this group, but mandated for patients with node-negative (N0) M0 disease only since November 2011. EXPOSURES Long-term HT either alone or with RT, as per local standard. Planned RT use was recorded at entry. MAIN OUTCOMES AND MEASURES Failure-free survival (FFS) and overall survival. RESULTS A total of 721 men with newly diagnosed M0 disease were included: median age at entry, 66 (interquartile range [IQR], 61-72) years, median (IQR) prostate-specific antigen level of 43 (18-88) ng/mL. There were 40 deaths (31 owing to prostate cancer) with 17 months' median follow-up. Two-year survival was 96% (95% CI, 93%-97%) and 2-year FFS, 77% (95% CI, 73%-81%). Median (IQR) FFS was 63 (26 to not reached) months. Time to FFS was worse in patients with N+ disease (HR, 2.02 [95% CI, 1.46-2.81]) than in those with N0 disease. Failure-free survival outcomes favored planned use of RT for patients with both N0M0 (HR, 0.33 [95% CI, 0.18-0.61]) and N+M0 disease (HR, 0.48 [95% CI, 0.29-0.79]). CONCLUSIONS AND RELEVANCE Survival for men entering the cohort with high-risk M0 disease was higher than anticipated at study inception. These nonrandomized data were consistent with previous trials that support routine use of RT with HT in patients with N0M0 disease. Additionally, the data suggest that the benefits of RT extend to men with N+M0 disease. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00268476; ISRCTN78818544.
Collapse
Affiliation(s)
- Nicholas D James
- Warwick Medical School, University of Warwick, Coventry, United Kingdom2University Hospitals Birmingham NHS Foundation Trust, The Medical School, University of Birmingham, Birmingham, United Kingdom
| | - Melissa R Spears
- Medical Research Council Clinical Trials Unit, University College, London, United Kingdom
| | - Noel W Clarke
- Department of Urology, Christie NHS Foundation Trust, Manchester, United Kingdom
| | - David P Dearnaley
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, United Kingdom
| | | | - Christopher C Parker
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Alastair W S Ritchie
- Medical Research Council Clinical Trials Unit, University College, London, United Kingdom
| | - J Martin Russell
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Francesca Schiavone
- Medical Research Council Clinical Trials Unit, University College, London, United Kingdom
| | - Gerhardt Attard
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Johann S de Bono
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, United Kingdom
| | | | | | - Tony Elliott
- Greater Manchester Group, Manchester, United Kingdom
| | | | | | | | | | - Jan Wallace
- Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Jim Barber
- Velindre Hospital, Cardiff, United Kingdom
| | | | - Mahesh K B Parmar
- Medical Research Council Clinical Trials Unit, University College, London, United Kingdom
| | - Matthew R Sydes
- Medical Research Council Clinical Trials Unit, University College, London, United Kingdom
| |
Collapse
|
158
|
Müller AC, Eckert F, Paulsen F, Zips D, Stenzl A, Schilling D, Alber M, Bares R, Martus P, Weckermann D, Belka C, Ganswindt U. Nodal Clearance Rate and Long-Term Efficacy of Individualized Sentinel Node–Based Pelvic Intensity Modulated Radiation Therapy for High-Risk Prostate Cancer. Int J Radiat Oncol Biol Phys 2016; 94:263-71. [DOI: 10.1016/j.ijrobp.2015.10.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/10/2015] [Accepted: 10/20/2015] [Indexed: 11/28/2022]
|
159
|
Mitchell DL, Tracy CR, Buatti JM, Smith MC, Snow AN, Henry MD, Vaena DA, Tewfik HH, Watkins JM. Individualization of Adjuvant Therapy After Radical Prostatectomy for Clinically Localized Prostate Cancer: Current Status and Future Directions. Clin Genitourin Cancer 2016; 14:12-21. [PMID: 26341039 DOI: 10.1016/j.clgc.2015.07.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 07/30/2015] [Indexed: 11/20/2022]
Abstract
Radiation therapy indications in the postprostatectomy setting are evolving. Several retrospective series have identified a number of "high-risk" pathologic features associated with an elevated risk of disease recurrence after radical prostatectomy. More recently, several randomized phase III trials demonstrated superior biochemical relapse-free survival for adjuvant radiation therapy after prostatectomy for patients with these high-risk pathologic features, including positive margin status, extraprostatic extension, and/or seminal vesicle invasion. These series further suggested improvement in distant metastasis control and overall survival after 15 years. However, not all patients with high-risk features experience disease recurrence after surgery alone, and some subsets of patients experience suboptimal disease control and survival despite immediate postoperative radiotherapy. Furthermore, some patients without high-risk features will develop recurrence. The present review discusses the current data and potential future directions to improve individualization of therapy after prostatectomy.
Collapse
Affiliation(s)
- Darrion L Mitchell
- Department of Radiation Oncology, University of Iowa Carver School of Medicine, Iowa City, IA
| | - Chad R Tracy
- Department of Urology, University of Iowa Carver School of Medicine, Iowa City, IA
| | - John M Buatti
- Department of Radiation Oncology, University of Iowa Carver School of Medicine, Iowa City, IA
| | - Mark C Smith
- Department of Radiation Oncology, University of Iowa Carver School of Medicine, Iowa City, IA
| | - Anthony N Snow
- Department of Pathology, University of Iowa Carver School of Medicine, Iowa City, IA
| | - Michael D Henry
- Department of Molecular Physiology and Biophysics and Holden Comprehensive Cancer Center, University of Iowa Carver School of Medicine, Iowa City, IA
| | - Daniel A Vaena
- Department of Hematology and Oncology, University of Iowa Carver School of Medicine, Iowa City, IA
| | | | - John M Watkins
- Department of Radiation Oncology, University of Iowa Carver School of Medicine, Iowa City, IA.
| |
Collapse
|
160
|
The number of risk factors is the strongest predictor of prostate cancer mortality: multi-institutional outcomes of an extreme-risk prostate cancer cohort. Clin Transl Oncol 2016; 18:1026-33. [PMID: 26781470 DOI: 10.1007/s12094-016-1481-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 01/04/2016] [Indexed: 10/22/2022]
Abstract
PURPOSE To report treatment outcomes in a cohort of extreme-risk prostate cancer patients and identify a subgroup of patients with worse prognosis. MATERIALS AND METHODS Extreme-risk prostate cancer patients were defined as patients with at least one extreme-risk factor: stage cT3b-cT4, Gleason score 9-10 or PSA > 50 ng/ml; or patients with 2 or more high-risk factors: stage cT2c-cT3a, Gleason 8 and PSA > 20 ng/ml. Overall survival (OS), cause-specific survival (CSS), clinical-free survival (CFS), and biochemical non-evidence of disease (bNED) survival are the four outcomes of interest in a population of 1341 patients. RESULTS With a median follow-up of 71.5 months, 5- and 10-year bNED survival, CFS, CSS and OS for the entire cohort were 77.1 % and 57.0, 89.2 and 78.9 %, 97.4 and 93.6 %, and 92.0 and 71.3 %, respectively. On multivariate analysis, PSA and clinical stage were associated with bNED survival. PSA and Gleason score predicted for CFS, whereas only Gleason score predicted for OS. When a simplified model was performed using the "number of risk factors" variable, this model provided the best distinction between patients with ≥2 extreme-risk factors and patients with 2 high-risk factors, showing a hazard ratio (HR) of 1.737 (p = 0.0003) for bNED survival, HR 1.743 (p = 0.0448) for OS and an HR of 3.963 (p = 0.0039) for the CSS endpoint. CONCLUSIONS Patients presenting at diagnosis with two extreme-risk criteria have almost fourfold higher risk for prostate cancer mortality. Such patients should be considered for more aggressive multimodal treatments.
Collapse
|
161
|
Chen CP, Johnson J, Seo Y, Weinberg VK, Shinohara K, Hsu ICJ, Roach M. Sentinel lymph node imaging guided IMRT for prostate cancer: Individualized pelvic radiation therapy versus RTOG guidelines. Adv Radiat Oncol 2016; 1:51-58. [PMID: 28799574 PMCID: PMC5506713 DOI: 10.1016/j.adro.2015.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/24/2015] [Accepted: 11/30/2015] [Indexed: 11/27/2022] Open
Abstract
Purpose/Objectives Current Radiation Therapy Oncology Group (RTOG) guidelines for pelvic radiation therapy are based on general anatomic boundaries. Sentinel lymph node (SLN) imaging can identify potential sites of lymph node involvement. We sought to determine how tailored radiation therapy fields for prostate cancer would compare to standard RTOG-based fields. Such individualized radiation therapy could prioritize the most important areas to irradiate while potentially avoiding coverage in areas where critical structures would be overdosed. Individualized radiation therapy could therefore increase the therapeutic index of pelvic radiation therapy. Methods and materials Ten intermediate or high-risk prostate cancer patients received androgen deprivation therapy with definitive radiation therapy, including an SLN imaging–tailored elective nodal volume (ENV). For dosimetric analyses, the ENV was recontoured using RTOG guidelines (RTOG_ENV) and on SLNs alone (SLN_ENV). Separate intensity modulated radiation therapy (IMRT) plans were optimized using RTOG_ENV and SLN_ENV for each patient. Dosimetric comparisons for these IMRT plans were performed for each patient. Dose differences to targets and critical structures among the different IMRT plans were calculated. Distributions of dose parameters were analyzed using non-parametric methods. Results Sixty percent of patients had SLNs outside of the RTOG_ENV. The larger volume IMRT plans covering SLN imaging–tailored elective nodal volume exhibited no significant dose differences versus plans covering RTOG_ENV. IMRT plans covering only the SLNs had significantly lower doses to bowel and femoral heads. Conclusions SLN-guided pelvic radiation therapy can be used to either treat the most critical nodes only or as an addition to RTOG guided pelvic radiation therapy to ensure that the most important nodes are included.
Collapse
|
162
|
Amini A, Kavanagh BD, Rusthoven CG. Improved survival with the addition of radiotherapy to androgen deprivation: questions answered and a review of current controversies in radiotherapy for non-metastatic prostate cancer. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:14. [PMID: 26855950 PMCID: PMC4716946 DOI: 10.3978/j.issn.2305-5839.2015.10.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 09/25/2015] [Indexed: 11/14/2022]
Abstract
The contemporary standard of care for locally advanced high-risk prostate cancer includes a combination of dose-escalated radiotherapy (RT) plus androgen-deprivation therapy (ADT). However, 20 years ago, at the inception of the National Cancer Institute of Canada (NCIC) led study (NCIC Clinical Trials Group PR.3/Medical Research Council PR07/Intergroup T94-0110), the survival impact of prostate RT for high-risk disease was uncertain. Recently, Mason, Warde and colleagues presented the final results of this NCIC/MRC study (PMID: 25691677) randomizing 1,205 high-risk prostate cancer patients to ADT + RT vs. ADT alone. These updated results confirm substantial improvements with the addition of RT to ADT for the endpoints of overall survival (OS), disease-free survival (DFS), and biochemical recurrence. Close examination of subtleties of this trial's design highlight some of the most salient controversies in the field of prostate RT, including the risk-stratified roles of ADT, optimal ADT duration, and RT field design in the dose-escalated and intensity-modulated radiotherapy (IMRT) era.
Collapse
Affiliation(s)
- Arya Amini
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Brian D Kavanagh
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Chad G Rusthoven
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
163
|
Jeong S, Lee JH, Chung MJ, Lee SW, Lee JW, Kang DG, Kim SH. Analysis of Geometric Shifts and Proper Setup-Margin in Prostate Cancer Patients Treated With Pelvic Intensity-Modulated Radiotherapy Using Endorectal Ballooning and Daily Enema for Prostate Immobilization. Medicine (Baltimore) 2016; 95:e2387. [PMID: 26765418 PMCID: PMC4718244 DOI: 10.1097/md.0000000000002387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
We evaluate geometric shifts of daily setup for evaluating the appropriateness of treatment and determining proper margins for the planning target volume (PTV) in prostate cancer patients.We analyzed 1200 sets of pretreatment megavoltage-CT scans that were acquired from 40 patients with intermediate to high-risk prostate cancer. They received whole pelvic intensity-modulated radiotherapy (IMRT). They underwent daily endorectal ballooning and enema to limit intrapelvic organ movement. The mean and standard deviation (SD) of daily translational shifts in right-to-left (X), anterior-to-posterior (Y), and superior-to-inferior (Z) were evaluated for systemic and random error.The mean ± SD of systemic error (Σ) in X, Y, Z, and roll was 2.21 ± 3.42 mm, -0.67 ± 2.27 mm, 1.05 ± 2.87 mm, and -0.43 ± 0.89°, respectively. The mean ± SD of random error (δ) was 1.95 ± 1.60 mm in X, 1.02 ± 0.50 mm in Y, 1.01 ± 0.48 mm in Z, and 0.37 ± 0.15° in roll. The calculated proper PTV margins that cover >95% of the target on average were 8.20 (X), 5.25 (Y), and 6.45 (Z) mm. Mean systemic geometrical shifts of IMRT were not statistically different in all transitional and three-dimensional shifts from early to late weeks. There was no grade 3 or higher gastrointestinal or genitourianry toxicity.The whole pelvic IMRT technique is a feasible and effective modality that limits intrapelvic organ motion and reduces setup uncertainties. Proper margins for the PTV can be determined by using geometric shifts data.
Collapse
Affiliation(s)
- Songmi Jeong
- From the Department of Radiation Oncology, St. Vincent Hospital, College of Medicine, The Catholic University of Korea, Suwon (SJ, JHL, MJC, SWL, DGK, SHK); and Department of Radiation Oncology, Kyungpook National University Hospital, Daegu, Korea (JWL)
| | | | | | | | | | | | | |
Collapse
|
164
|
Guidance on patient consultation. Current evidence for prostate-specific antigen screening in healthy men and treatment options for men with proven localised prostate cancer. Curr Urol Rep 2015; 16:28. [PMID: 25773347 DOI: 10.1007/s11934-015-0502-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The main objective of this review is to summarise, for primary and secondary care doctors, the management options and current supporting evidence for clinically localised prostate cancer. We review all aspects of management including current guidelines on early cancer detection and the importance of informed consent on PSA-based screening and assess the most common treatment options and the evidence for managing patients with low-, medium-, and high-risk disease.
Collapse
|
165
|
Whole Pelvis Versus Prostate-Only Radiotherapy With or Without Short-Course Androgen Deprivation Therapy and Mortality Risk. Clin Genitourin Cancer 2015; 13:555-61. [DOI: 10.1016/j.clgc.2015.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 04/23/2015] [Accepted: 04/26/2015] [Indexed: 11/18/2022]
|
166
|
Williams S. Diagnosis and management of locally advanced disease. Prostate Int 2015. [DOI: 10.1016/j.prnil.2015.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
167
|
Sargos P, Guerif S, Latorzeff I, Hennequin C, Pommier P, Lagrange JL, Créhange G, Chapet O, de Crevoisier R, Azria D, Supiot S, Habibian M, Soulié M, Richaud P. Definition of lymph node areas for radiotherapy of prostate cancer: A critical literature review by the French Genito-Urinary Group and the French Association of Urology (GETUG-AFU). Cancer Treat Rev 2015; 41:814-20. [DOI: 10.1016/j.ctrv.2015.10.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 10/15/2015] [Accepted: 10/18/2015] [Indexed: 12/30/2022]
|
168
|
Wu R, Woodford H, Capp A, Hunter P, Cowin G, Tai KH, Nguyen PL, Chong P, Martin J. A prospective study of nomogram-based adaptation of prostate radiotherapy target volumes. Radiat Oncol 2015; 10:243. [PMID: 26607977 PMCID: PMC4660680 DOI: 10.1186/s13014-015-0545-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 11/15/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A prospective clinical trial was conducted to evaluate the feasibility of a novel approach to the treatment of patients with high risk prostate cancer (HRPC) through the use of a nomogram to tailor radiotherapy target volumes. METHODS Twenty seven subjects with HRPC were treated with a mildly hypofractionated radiotherapy regimen using image-guided IMRT technique between Jun/2013-Jan/2015. A set of validated prognostic factors were inputted into the Memorial-Sloan-Kettering Cancer Center (MSKCC) prostate cancer nomogram to estimate risk of loco-regional spread (LRS). The nomogram risk estimates for extra-capsular extension (ECE), seminal vesicles involvement (SVI), and pelvic lymph nodes involvement (LNI) were used to adapt radiotherapy treatment volumes based on a risk threshold of ≥15 % in all cases. A planning guide was used to delineate target volumes and organs at risk (OAR). Up to three dose levels were administered over 28 fractions; 70Gy for gross disease in the prostate +/- seminal vesicles (2.5Gy/fraction), 61.6Gy for subclinical peri-prostatic disease (2.2Gy/fraction) and 50.4Gy to pelvic nodes (1.8Gy/fraction). Data regarding protocol adherence, nomogram use, radiotherapy dose distribution, and acute toxicity were collected. RESULTS Nomogram use 100 % of patients were treated for ECE, 88.9 % for SVI, and 70.4 % for LNI. The three areas at risk of LRS were appropriately treated according to the study protocol in 98.8 % cases. The MSKCC nomogram estimates for LRS differed significantly between the time of recruitment and analysis. Contouring protocol compliance Compliance with the trial contouring protocol for up to seven target volumes was 93.0 % (159/171). Compliance with protocol for small bowel contouring was poor (59.3 %). Dose constraints compliance Compliance with dose constraints for target volumes was 97.4 % (191/196). Compliance with dose constraints for OAR was 88.2 % (285/323). Acute toxicity There were no grade 3 acute toxicities observed. 20/27 (74.1 %) and 6/27 (22.2 %) patients experienced a grade 2 genitourinary and gastrointestinal toxicity respectively. CONCLUSIONS We have demonstrated the feasibility of this novel risk-adapted radiation treatment protocol for HRPC. This study has identified key learning points regarding this approach, including the importance of standardization and updating of risk quantification tools, and the utility of an observer to verify their correct use. TRIAL REGISTRATION ClincialTrials.gov identifier NCT01418040 . Hunter New England Human Research Ethics Committee (HNEHREC) reference number 12/08/15/4.02.
Collapse
Affiliation(s)
- Raymond Wu
- Department of Radiation Oncology, Calvary Mater Newcastle, Edith Street, Waratah, NSW, 2298, Australia.
| | - Hannah Woodford
- University of Newcastle, School of Medicine and Public Health, University Drive, Callaghan, NSW, 2308, Australia.
| | - Anne Capp
- Department of Radiation Oncology, Calvary Mater Newcastle, Edith Street, Waratah, NSW, 2298, Australia.
| | - Perry Hunter
- Department of Radiation Oncology, Calvary Mater Newcastle, Edith Street, Waratah, NSW, 2298, Australia.
| | - Gary Cowin
- University of Queensland, The Centre for Advanced Imaging, Building 57, Research Road, St Lucia, QLD, 4072, Australia.
| | - Keen-Hun Tai
- Department of Radiation Oncology, Peter MacCallum Cancer Institute, St Andrews Place, East Melbourne, VIC, 3002, Australia.
| | - Paul L Nguyen
- Brigham and Women's Hospital, Radiation Oncology, 75 Francis Street, Boston, MA, 02115, USA.
| | - Peter Chong
- Sky Central East, Level 3, Suite 2, 20 Smart Street, Charlestown, NSW, 2290, Australia.
| | - Jarad Martin
- Department of Radiation Oncology, Calvary Mater Newcastle, Edith Street, Waratah, NSW, 2298, Australia. .,University of Newcastle, School of Medicine and Public Health, University Drive, Callaghan, NSW, 2308, Australia. .,University of Queensland, The Centre for Advanced Imaging, Building 57, Research Road, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
169
|
|
170
|
Lilleby W, Narrang A, Tafjord G, Vlatkovic L, Russnes KM, Stensvold A, Hole KH, Tran P, Eilertsen K. Favorable outcomes in locally advanced and node positive prostate cancer patients treated with combined pelvic IMRT and androgen deprivation therapy. Radiat Oncol 2015; 10:232. [PMID: 26577452 PMCID: PMC4650510 DOI: 10.1186/s13014-015-0540-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/10/2015] [Indexed: 11/22/2022] Open
Abstract
Background The most appropriate treatment for men with prostate cancer and positive pelvic nodes, N+, is an area of active controversy. We report our 5-years outcomes in men with locally advanced prostate cancer (T1-T4N0-N1M0) treated with definitive radiotherapy encompassing the prostate and pelvic lymph nodes (intensity modulated radiotherapy, IMRT) and long-term androgen deprivation therapy (ADT). Material and methods Of the 138 consecutive eligible men all living patients have been followed up to almost 5 years. Survival endpoints for 5-year biochemical failure-free survival (BFFS), relapse-free survival (RFS), prostate cancer-specific survival (PCSS), and overall survival (OS) were assessed by Kaplan-Meier analysis. Univariate and multivariate Cox regression proportional hazards models were constructed for all survival endpoints. The RTOG morbidity grading system for physician rated toxicity was applied. Results Patients with locally advanced T3-T4 tumors (35 %) and N1 (51 %) have favorable outcome when long-term ADT is combined with definitive radiotherapy encompassing pelvic lymph nodes. The 5-year BFFS, RFS, PCSS and OS were 71.4, 76.2, 94.5 and 89.0 %, respectively. High Gleason sum (9–10) had a strong independent prognostic impact on BFFS, RFS and OS (p = 0.001, <0.001, and 0.005 respectively). The duration of ADT (= > 28 months) showed a significant independent association with improved PCSS (p = 0.02) and OS (p = 0.001). Lymph node involvement was not associated with survival endpoints in the multivariate analysis. The radiotherapy induced toxicity seen in our study population was moderate with rare Grade 3 GI side effects and up to 11 % for Grade 3 GU consisting mainly of urgency and frequency. Conclusion Pelvic IMRT in combination with long-term ADT can achieve long-lasting disease control in men with N+ disease and unfavorable prognostic factors. Electronic supplementary material The online version of this article (doi:10.1186/s13014-015-0540-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wolfgang Lilleby
- Department of Oncology, Oslo University Hospital, The Norwegian Radium Hospital, 0424, Oslo, Norway.
| | - Amol Narrang
- Departments of Radiation Oncology and Molecular Radiation Sciences, Oncology and Urology, Johns Hopkins Hospital, Baltimore, MD, USA.
| | - Gunnar Tafjord
- Department of Oncology, Oslo University Hospital, The Norwegian Radium Hospital, 0424, Oslo, Norway.
| | - Ljiljana Vlatkovic
- Department of Pathology, Oslo University Hospital, The Norwegian Radium Hospital, 0424, Oslo, Norway.
| | - Kjell Magne Russnes
- Department of Oncology, Oslo University Hospital, The Norwegian Radium Hospital, 0424, Oslo, Norway.
| | - Andreas Stensvold
- Department of Oncology, Østfold Hospital Trust, 1603, Fredrikstad, Norway.
| | - Knut Håkon Hole
- Department of Radiology, Oslo University Hospital, The Norwegian Radium Hospital, 0424, Oslo, Norway.
| | - Phuoc Tran
- Departments of Radiation Oncology and Molecular Radiation Sciences, Oncology and Urology, Johns Hopkins Hospital, Baltimore, MD, USA.
| | - Karsten Eilertsen
- Department of Medical Physics, Oslo University Hospital, The Norwegian Radium Hospital, 0424, Oslo, Norway.
| |
Collapse
|
171
|
Habl G, Katayama S, Uhl M, Kessel KA, Edler L, Debus J, Herfarth K, Sterzing F. Helical intensity-modulated radiotherapy of the pelvic lymph nodes with a simultaneous integrated boost to the prostate--first results of the PLATIN 1 trial. BMC Cancer 2015; 15:868. [PMID: 26547188 PMCID: PMC4637144 DOI: 10.1186/s12885-015-1886-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 10/30/2015] [Indexed: 11/10/2022] Open
Abstract
Background Definitive, percutaneous irradiation of the prostate and the pelvic lymph nodes in high-risk prostate cancer is the alternative to prostatectomy plus lymphadenectomy. To date, the role of whole pelvis radiotherapy (WPRT) has not been clarified especially taking into consideration the benefits of high conformal IMRT (intensity modulated radiotherapy) of complex-shaped target volumes. Methods From 2009 to 2012, 40 patients of high-risk prostate cancer with an increased risk of microscopic lymph node involvement were enrolled into this prospective phase II trial. Patients received at least two months of antihormonal treatment (AT) before radiotherapy continuing for at least 2 years. Helical IMRT (tomotherapy) of the pelvic lymph nodes (51.0 Gy) with a simultaneous integrated, moderate hypofractionated boost (single dose of 2.25 Gy) to the prostate (76.5 Gy) was performed in 34 fractions. PSA levels, prostate-related symptoms and quality of life were assessed at regular intervals for 24 months. Results Of the 40 patients enrolled, 38 finished the treatment as planned. Overall acute toxicity rates were low and no acute grade 3 or 4 gastrointestinal (GI) and genitourinary (GU) toxicity occurred. 21.6 % of patients experienced acute grade 2 but no late grade ≥2 GI toxicity. Regarding GU side effects, results showed 48.6 % acute grade 2 and 6.4 % late grade 2 toxicity. After a median observation time of 23.4 months the PLATIN 1 trial can be considered as sufficiently safe meeting the prospectively defined aims of the trial. With 34/37 patients free of a PSA recurrence it shows promising efficacy. Conclusion Tomotherapy of the pelvic lymph nodes with a simultaneous integrated boost to the prostate can be performed safely and without excessive toxicity. The combined irradiation of both prostate and pelvic lymph nodes seems to be as well tolerated as the irradiation of the prostate alone. Trial registration Trial Numbers: ARO 2009–05, ClinicalTrials.gov: NCT01903408.
Collapse
Affiliation(s)
- Gregor Habl
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany. .,Department of Radiation Oncology, Technische Universität München (TUM), Munich, Germany.
| | - Sonja Katayama
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Matthias Uhl
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Kerstin A Kessel
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,Department of Radiation Oncology, Technische Universität München (TUM), Munich, Germany
| | - Lutz Edler
- Department of Biostatistics, German Cancer Research Center, Heidelberg, Germany
| | - Juergen Debus
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Klaus Herfarth
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Florian Sterzing
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| |
Collapse
|
172
|
Bhattacharyya T, Purushothaman K, Puthiyottil SSV, Muttath G. External Beam Radiation Therapy and Abiraterone in Men With Localized Prostate Cancer: Safety and Effect on Tissue Androgens. In Regard to Cho et al. Int J Radiat Oncol Biol Phys 2015; 93:719. [PMID: 26461012 DOI: 10.1016/j.ijrobp.2015.06.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 06/16/2015] [Indexed: 10/23/2022]
Affiliation(s)
- Tapesh Bhattacharyya
- Department of Radiation Oncology, Malabar Cancer Centre, Thalassery, Kerala, India
| | - Kiran Purushothaman
- Department of Radiation Oncology, Malabar Cancer Centre, Thalassery, Kerala, India
| | | | - Geetha Muttath
- Department of Radiation Oncology, Malabar Cancer Centre, Thalassery, Kerala, India
| |
Collapse
|
173
|
Spratt DE, Evans MJ, Davis BJ, Doran MG, Lee MX, Shah N, Wongvipat J, Carnazza KE, Klee GG, Polkinghorn W, Tindall DJ, Lewis JS, Sawyers CL. Androgen Receptor Upregulation Mediates Radioresistance after Ionizing Radiation. Cancer Res 2015; 75:4688-96. [PMID: 26432404 DOI: 10.1158/0008-5472.can-15-0892] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 08/11/2015] [Indexed: 12/19/2022]
Abstract
Clinical trials have established the benefit of androgen deprivation therapy (ADT) combined with radiotherapy in prostate cancer. ADT sensitizes prostate cancer to radiotherapy-induced death at least in part through inhibition of DNA repair machinery, but for unknown reasons, adjuvant ADT provides further survival benefits. Here, we show that androgen receptor (AR) expression and activity are durably upregulated following radiotherapy in multiple human prostate cancer models in vitro and in vivo. Moreover, the degree of AR upregulation correlates with survival in vitro and time to tumor progression in animal models. We also provide evidence of AR pathway upregulation, measured by a rise in serum levels of AR-regulated hK2 protein, in nearly 20% of patients after radiotherapy. Furthermore, these men were three-fold more likely to experience subsequent biochemical failure. Collectively, these data demonstrate that radiotherapy can upregulate AR signaling after therapy to an extent that negatively affects disease progression and/or survival.
Collapse
Affiliation(s)
- Daniel E Spratt
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York. Human Oncology Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Radiology and Molecular the Molecular Pharmacology & Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael J Evans
- Human Oncology Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brian J Davis
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Michael G Doran
- Department of Radiology and Molecular the Molecular Pharmacology & Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Man Xia Lee
- Human Oncology Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Neel Shah
- Human Oncology Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John Wongvipat
- Human Oncology Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kathryn E Carnazza
- Department of Radiology and Molecular the Molecular Pharmacology & Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - George G Klee
- Departments of Urology and Biochemistry/Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - William Polkinghorn
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York. Human Oncology Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Donald J Tindall
- Departments of Urology and Biochemistry/Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Jason S Lewis
- Department of Radiology and Molecular the Molecular Pharmacology & Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Charles L Sawyers
- Human Oncology Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
174
|
Barbosa Neto O, Souhami L, Faria S. Hypofractionated radiation therapy for prostate cancer: The McGill University Health Center experience. Cancer Radiother 2015; 19:431-6. [DOI: 10.1016/j.canrad.2015.05.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 05/19/2015] [Accepted: 05/20/2015] [Indexed: 10/23/2022]
|
175
|
Blanchard P, Faivre L, Lesaunier F, Salem N, Mesgouez-Nebout N, Deniau-Alexandre E, Rolland F, Ferrero JM, Houédé N, Mourey L, Théodore C, Krakowski I, Berdah JF, Baciuchka M, Laguerre B, Davin JL, Habibian M, Culine S, Laplanche A, Fizazi K. Outcome According to Elective Pelvic Radiation Therapy in Patients With High-Risk Localized Prostate Cancer: A Secondary Analysis of the GETUG 12 Phase 3 Randomized Trial. Int J Radiat Oncol Biol Phys 2015; 94:85-92. [PMID: 26576711 DOI: 10.1016/j.ijrobp.2015.09.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 09/04/2015] [Accepted: 09/15/2015] [Indexed: 11/29/2022]
Abstract
PURPOSE The role of pelvic elective nodal irradiation (ENI) in the management of prostate cancer is controversial. This study analyzed the role of pelvic radiation therapy (RT) on the outcome in high-risk localized prostate cancer patients included in the Groupe d'Etude des Tumeurs Uro-Genitales (GETUG) 12 trial. METHODS AND MATERIALS Patients with a nonpretreated high-risk localized prostate cancer and a staging lymphadenectomy were randomly assigned to receive either goserelin every 3 months for 3 years and 4 cycles of docetaxel plus estramustine or goserelin alone. Local therapy was administered 3 months after the start of systemic treatment. Performance of pelvic ENI was left to the treating physician. Only patients treated with primary RT were included in this analysis. The primary endpoint was biochemical progression-free survival (bPFS). RESULTS A total of 413 patients treated from 2002 to 2006 were included, of whom 358 were treated using primary RT. A total of 208 patients received pelvic RT and 150 prostate-only RT. Prostate-specific antigen (PSA) concentration, Gleason score, or T stage did not differ according to performance of pelvic RT; pN+ patients more frequently received pelvic RT than pN0 patients (P<.0001). Median follow-up was 8.8 years. In multivariate analysis, bPFS was negatively impacted by pN stage (hazard ratio [HR]: 2.52 [95% confidence interval [CI]: 1.78-3.54], P<.0001), Gleason score 8 or higher (HR: 1.41 [95% CI: 1.03-1.93], P=.033) and PSA higher than 20 ng/mL (HR: 1.41 [95% CI: 1.02-1.96], P=.038), and positively impacted by the use of chemotherapy (HR: 0.66 [95% CI: 0.48-0.9], P=.009). There was no association between bPFS and use of pelvic ENI in multivariate analysis (HR: 1.10 [95% CI: 0.78-1.55], P=.60), even when analysis was restricted to pN0 patients (HR: 0.88 [95% CI: 0.59-1.31], P=.53). Pelvic ENI was not associated with increased acute or late patient reported toxicity. CONCLUSIONS This unplanned analysis of a randomized trial failed to demonstrate a benefit of pelvic ENI on bPFS in high-risk localized prostate cancer patients.
Collapse
Affiliation(s)
- Pierre Blanchard
- Radiation Oncology, Gustave Roussy Cancer Center, Villejuif, France; University of Paris-Sud, Cancer Campus, Villejuif, France.
| | - Laura Faivre
- Biostatistics, Gustave Roussy Cancer Center, Villejuif, France
| | | | - Naji Salem
- Radiation Oncology, Institut Paoli Calmette, Marseille, France
| | | | | | - Frédéric Rolland
- Medical Oncology, Institut de Cancérologie de l'Ouest, Nantes, France
| | | | - Nadine Houédé
- Medical Oncology, Institut Bergonié, Bordeaux, France
| | | | | | | | | | | | | | | | | | - Stéphane Culine
- Department of Medical Oncology, Hopital Saint-Louis, APHP, Paris, France
| | - Agnès Laplanche
- Biostatistics, Gustave Roussy Cancer Center, Villejuif, France
| | - Karim Fizazi
- University of Paris-Sud, Cancer Campus, Villejuif, France; Department of Cancer Medicine, Gustave Roussy Cancer Center, Villejuif, France
| |
Collapse
|
176
|
Amini A, Jones BL, Yeh N, Rusthoven CG, Armstrong H, Kavanagh BD. Survival Outcomes of Whole-Pelvic Versus Prostate-Only Radiation Therapy for High-Risk Prostate Cancer Patients With Use of the National Cancer Data Base. Int J Radiat Oncol Biol Phys 2015; 93:1052-63. [PMID: 26581142 DOI: 10.1016/j.ijrobp.2015.09.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 08/25/2015] [Accepted: 09/08/2015] [Indexed: 11/27/2022]
Abstract
PURPOSE/OBJECTIVES The addition of whole pelvic (WP) compared with prostate-only (PO) radiation therapy (RT) for clinically node-negative prostate cancer remains controversial. The purpose of our study was to evaluate the survival benefit of adding WPRT versus PO-RT for high-risk, node-negative prostate cancer, using the National Cancer Data Base (NCDB). METHODS AND MATERIALS Patients with high-risk prostate cancer treated from 2004 to 2006, with available data for RT volume, coded as prostate and pelvis (WPRT) or prostate alone (PO-RT) were included. Multivariate analysis (MVA) and propensity-score matched analysis (PSM) were performed. Recursive partitioning analysis (RPA) based on overall survival (OS) using Gleason score (GS), T stage, and pretreatment prostate-specific antigen (PSA) was also conducted. RESULTS A total of 14,817 patients were included: 7606 (51.3%) received WPRT, and 7211 (48.7%) received PO-RT. The median follow-up time was 81 months (range, 2-122 months). Under MVA, the addition of WPRT for high-risk patients had no OS benefit compared with PO-RT (HR 1.05; P=.100). On subset analysis, patients receiving dose-escalated RT also did not benefit from WPRT (HR 1.01; P=.908). PSM confirmed no survival benefit with the addition of WPRT for high-risk patients (HR 1.05; P=.141). In addition, RPA was unable to demonstrate a survival benefit of WPRT for any subset. Other prognostic factors for inferior OS under MVA included older age (HR 1.25; P<.001), increasing comorbidity scores (HR 1.46; P<.001), higher T stage (HR 1.17; P<.001), PSA (HR 1.81; P<.001), and GS (HR 1.29; P<.001), and decreasing median county household income (HR 1.15; P=.011). Factors improving OS included the addition of androgen deprivation therapy (HR 0.92; P=.033), combination external beam RT plus brachytherapy boost (HR 0.71; P<.001), and treatment at an academic/research institution (HR 0.84; P=.002). CONCLUSION In the largest reported analysis of WPRT for patients with high-risk prostate cancer treated in the dose-escalated era, the addition of WPRT demonstrated no survival advantage compared with PO-RT.
Collapse
Affiliation(s)
- Arya Amini
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, Colorado
| | - Bernard L Jones
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, Colorado
| | - Norman Yeh
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, Colorado
| | - Chad G Rusthoven
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, Colorado
| | - Hirotatsu Armstrong
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, Colorado
| | - Brian D Kavanagh
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, Colorado.
| |
Collapse
|
177
|
Liu HX, Du L, Yu W, Cai BN, Xu SP, Xie CB, Ma L. Hypofractionated Helical Tomotherapy for Older Aged Patients With Prostate Cancer: Preliminary Results of a Phase I-II Trial. Technol Cancer Res Treat 2015; 15:546-54. [PMID: 26152749 DOI: 10.1177/1533034615593189] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 05/28/2015] [Indexed: 11/16/2022] Open
Abstract
In our center, the feasibility and related acute toxicities of hypofractionated helical tomotherapy have been evaluated in older aged patients with prostate cancer . Between February 2009 and February 2014, 67 patients (older than 65 years) were enrolled in a prospective phase I-II study (registered number, ChiCTR-ONC-13004037). Patients in cohort 1 (n = 33) and cohort 2 (n = 34) received 76 Gy in 34 fractions (2.25 Gy/F) and 71.6 Gy in 28 fractions (2.65 Gy/F), respectively, to the prostate and seminal vesicles, while 25 patients in cohort 2 also received integrated elective lymph node irradiation (50.4 Gy). All patients were treated with helical tomotherapy, and daily image guidance was performed before each treatment. Acute toxicities were assessed with Radiation Therapy Oncology Group (RTOG)/European Organization for Research on Treatment of Cancer (EORTC) criteria. No significant difference was detected between the 2 cohorts in the incidence of acute toxicities. In cohort 1, the incidences of grade 1 and 2 genitourinary and gastrointestinal toxicities were 45.5% and 45.4%, respectively, and without grade 3 and 4 toxicities. In cohort 2, the incidences of acute grade 1 and 2 genitourinary and gastrointestinal toxicities were 47.1% and 55.9%, respectively, and grade 3 genitourinary toxicity (hematuria) was noted only in 1 patient. No significant difference was detected in the incidence of acute toxicities between the patients receiving integrated elective lymph node irradiation and those receiving irradiation to prostate and seminal vesicle in cohort 2. Univariate and multivariate analyses were performed with clinical parameters. Only the baseline weight was found negatively correlated with genitourinary toxicities at a weak level (relative risk = 0.946, 95% confidence interval 0.896-0.998], P = .043). This study shows that 2 hypofractionation regimens (76 Gy/34F and 71.6 Gy/28F) delivered with HT are well tolerated in older aged patients having prostate cancer without significant difference for acute toxicities between the 2 cohorts. Late toxicities and treatment outcomes for these patients are under investigation.
Collapse
Affiliation(s)
- Hai-Xia Liu
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, China
| | - Lei Du
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, China Department of Radiation Oncology, Hainan Branch of Chinese PLA General Hospital, Sanya, China
| | - Wei Yu
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, China
| | - Bo-Ning Cai
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, China
| | - Shou-Ping Xu
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, China
| | - Chuan-Bin Xie
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, China
| | - Lin Ma
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing, China Department of Radiation Oncology, Hainan Branch of Chinese PLA General Hospital, Sanya, China
| |
Collapse
|
178
|
Harris VA, Staffurth J, Naismith O, Esmail A, Gulliford S, Khoo V, Lewis R, Littler J, McNair H, Sadoyze A, Scrase C, Sohaib A, Syndikus I, Zarkar A, Hall E, Dearnaley D. Consensus Guidelines and Contouring Atlas for Pelvic Node Delineation in Prostate and Pelvic Node Intensity Modulated Radiation Therapy. Int J Radiat Oncol Biol Phys 2015; 92:874-83. [PMID: 26104940 DOI: 10.1016/j.ijrobp.2015.03.021] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/19/2015] [Accepted: 03/20/2015] [Indexed: 12/29/2022]
Abstract
PURPOSE The purpose of this study was to establish reproducible guidelines for delineating the clinical target volume (CTV) of the pelvic lymph nodes (LN) by combining the freehand Royal Marsden Hospital (RMH) and Radiation Therapy Oncology Group (RTOG) vascular expansion techniques. METHODS AND MATERIALS Seven patients with prostate cancer underwent standard planning computed tomography scanning. Four different CTVs (RMH, RTOG, modified RTOG, and Prostate and pelvIs Versus prOsTate Alone treatment for Locally advanced prostate cancer [PIVOTAL] trial) were created for each patient, and 6 different bowel expansion margins (BEM) were created to assess bowel avoidance by the CTV. The resulting CTVs were compared visually and by using Jaccard conformity indices. The volume of overlap between bowel and planning target volume (PTV) was measured to aid selection of an appropriate BEM to enable maximal LN yet minimal normal tissue coverage. RESULTS In total, 84 nodal contours were evaluated. LN coverage was similar in all groups, with all of the vascular-expansion techniques (RTOG, modified RTOG, and PIVOTAL), resulting in larger CTVs than that of the RMH technique (mean volumes: 287.3 cm(3), 326.7 cm(3), 310.3 cm(3), and 256.7 cm(3), respectively). Mean volumes of bowel within the modified RTOG PTV were 19.5 cm(3) (with 0 mm BEM), 17.4 cm(3) (1-mm BEM), 10.8 cm(3) (2-mm BEM), 6.9 cm(3) (3-mm BEM), 5.0 cm(3) (4-mm BEM), and 1.4 cm(3) (5-mm BEM) in comparison with an overlap of 9.2 cm(3) seen using the RMH technique. Evaluation of conformity between LN-CTVs from each technique revealed similar volumes and coverage. CONCLUSIONS Vascular expansion techniques result in larger LN-CTVs than the freehand RMH technique. Because the RMH technique is supported by phase 1 and 2 trial safety data, we proposed modifications to the RTOG technique, including the addition of a 3-mm BEM, which resulted in LN-CTV coverage similar to that of the RMH technique, with reduction in bowel and planning target volume overlap. On the basis of these findings, recommended guidelines including a detailed pelvic LN contouring atlas have been produced and implemented in the PIVOTAL trial.
Collapse
Affiliation(s)
- Victoria A Harris
- Academic Urology Unit, Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - John Staffurth
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, Wales, United Kingdom
| | - Olivia Naismith
- Joint Department of Physics, Institute of Cancer Research, and Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Alikhan Esmail
- Ipswich Hospital NHS Foundation Trust, Ipswich, United Kingdom
| | - Sarah Gulliford
- Joint Department of Physics, Institute of Cancer Research, and Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Vincent Khoo
- Academic Urology Unit, Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Rebecca Lewis
- Clinical Trials and Statistics Unit, Institute of Cancer Research, London, United Kingdom
| | - John Littler
- Clatterbridge Cancer Centre, Liverpool, United Kingdom
| | - Helen McNair
- Department of Radiotherapy, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Azmat Sadoyze
- Beatson West of Scotland Cancer Centre, Scotland, Glasgow, United Kingdom
| | | | - Aslam Sohaib
- Department of Radiology, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | | | - Anjali Zarkar
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Emma Hall
- Clinical Trials and Statistics Unit, Institute of Cancer Research, London, United Kingdom
| | - David Dearnaley
- Academic Urology Unit, Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, United Kingdom.
| |
Collapse
|
179
|
Zumsteg ZS, Spratt DE, Romesser PB, Pei X, Zhang Z, Kollmeier M, McBride S, Yamada Y, Zelefsky MJ. Anatomical Patterns of Recurrence Following Biochemical Relapse in the Dose Escalation Era of External Beam Radiotherapy for Prostate Cancer. J Urol 2015; 194:1624-30. [PMID: 26165583 DOI: 10.1016/j.juro.2015.06.100] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2015] [Indexed: 11/15/2022]
Abstract
PURPOSE We provide a comprehensive analysis of anatomical patterns of recurrence following external beam radiotherapy in patients with localized prostate cancer. MATERIALS AND METHODS This retrospective analysis included 2,694 patients with localized prostate cancer who received definitive, dose escalated external beam radiotherapy from 1991 to 2008. First recurrence sites were defined as initial sites of clinically detected recurrence and any subsequent clinically detected recurrence within 3 months. Anatomical recurrence patterns were classified as local (prostate/seminal vesicles only), lymphotropic (lymph nodes only) and osteotropic (bones only) in patients with disease confined only to these respective sites for at least 2 years from the initial clinically detected recurrence. RESULTS Prostate was the most common first recurrence site in the low, intermediate and high risk groups with an 8-year cumulative incidence of 3.5%, 9.8% and 14.6%, respectively. The 8-year risk of isolated pelvic lymph node relapse as the first recurrence site was 0%, 1.0% and 3.3%, respectively. In the 474 patients with clinically detected recurrence the most common first recurrence site was local in 55.3%, bone in 33.5%, pelvic lymph nodes in 21.3% and abdominal lymph nodes in 9.1%. Patients showed unique relapse distributions, including a pattern that was local in 41.6%, lymphotropic in 9.7%, osteotropic in 20.3% and multiorgan/visceral in 28.5%. Anatomical recurrence pattern was the strongest predictor of prostate cancer specific mortality on multivariate analysis of patients with clinically detected recurrence. CONCLUSIONS The most common first recurrence site after dose escalated external beam radiotherapy for prostate cancer is in the prostate and seminal vesicles in all risk groups. In contrast, patients treated without elective pelvic lymph node irradiation are at relatively low risk for isolated pelvic lymph node relapse. Recurrence patterns revealed a tropism for specific anatomical distributions with divergent prognoses, suggesting underlying biological differences among tumors.
Collapse
Affiliation(s)
- Zachary S Zumsteg
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California (ZSZ)
| | - Daniel E Spratt
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California (ZSZ)
| | - Paul B Romesser
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California (ZSZ)
| | - Xin Pei
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California (ZSZ)
| | - Zhigang Zhang
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California (ZSZ)
| | - Marisa Kollmeier
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California (ZSZ)
| | - Sean McBride
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California (ZSZ)
| | - Yoshiya Yamada
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California (ZSZ)
| | - Michael J Zelefsky
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California (ZSZ).
| |
Collapse
|
180
|
Defining the “Hostile Pelvis” for Intensity Modulated Radiation Therapy: The Impact of Anatomic Variations in Pelvic Dimensions on Dose Delivered to Target Volumes and Organs at Risk in Patients With High-Risk Prostate Cancer Treated With Whole Pelvic Radiation Therapy. Int J Radiat Oncol Biol Phys 2015; 92:894-903. [DOI: 10.1016/j.ijrobp.2015.03.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 03/14/2015] [Accepted: 03/17/2015] [Indexed: 11/18/2022]
|
181
|
Stereotactic Ablative Radiotherapy with CyberKnife in the Treatment of Locally Advanced Prostate Cancer: Preliminary Results. TUMORI JOURNAL 2015; 101:684-91. [DOI: 10.5301/tj.5000355] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2015] [Indexed: 11/20/2022]
Abstract
Aims and Background Recent clinical reports of stereotactic ablative radiotherapy (SABR) in the treatment of low-risk prostate cancer have been encouraging. Our study evaluates the efficacy and safety of SABR using the CyberKnife system for treating intermediate- to very-high-risk prostate cancer. Methods and Study Design Between May 2010 and June 2013, 31 patients (15 intermediate risk, 14 high risk, and 2 very high risk) without pelvic lymph node metastasis were enrolled retrospectively. The treatment consisted of 37.5 Gy in 5 fractions over 1-2 weeks using CyberKnife SABR. Twenty-five patients (81%) received androgen deprivation therapy (ADT). Biochemical failure was defined using the nadir + 2 criterion. Toxicity was assessed with the Common Terminology Criteria of Adverse Events (version 4). Results The median follow-up was 36 months (range 7-58 months). The median pretreatment prostate-pecific antigen (PSA) was 13.5 ng/mL (range 4.5-124.0 ng/mL). The median PSA decreased to 0.09 ng/mL (range <0.04-5.38 ng/mL) and 0.12 ng/mL (range <0.04-2.63 ng/mL) at 6 months and 12 months after SABR, respectively. The 3-year biochemical relapse-free survival was 90.2% for all patients, 100% for the intermediate-risk patients, and 82% for the high- and very-high-risk patients (p = 0.186). No patient experienced ≥ grade 3 toxicity. There were 7 acute and 5 late grade 2 genitourinary toxicities and 1 acute and no late grade 2 gastrointestinal toxicity. Conclusions Our preliminary results support that CyberKnife SABR with ADT is safe and feasible in patients with intermediate- to high-risk prostate cancer. A further large-scale clinical trial with longer follow-up is warranted.
Collapse
|
182
|
Okubo M, Nakayama H, Itonaga T, Tajima YU, Shiraishi S, Mikami R, Sakurada A, Sugahara S, Koizumi K, Tokuuye K. Impact of the duration of hormonal therapy following radiotherapy for localized prostate cancer. Oncol Lett 2015; 10:255-259. [PMID: 26171009 DOI: 10.3892/ol.2015.3216] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Accepted: 01/08/2015] [Indexed: 11/06/2022] Open
Abstract
External-beam radiotherapy (EBRT) combined with androgen deprivation therapy (ADT) is known to provide improved survival outcomes compared with EBRT alone in the treatment of prostate cancer; however, the use of ADT has been reported to be associated with adverse events. Accordingly, the aim of the present study was to clarify the adequate duration of ADT when combined with EBRT to treat patients with high-risk localized prostate cancer, with consideration of survival outcomes and toxicity. Between 2001 and 2011, 173 patients with high-risk localized prostate cancer received ADT combined with EBRT, at a median dose of 69.6 Gy. Of these, 54 (31%) underwent short-term ADT (<36 months) and 119 (69%) underwent long-term ADT (≥36 months). During the median follow-up period of 54 months, the five-year progression-free survival rate of patients receiving short-term ADT (72.9%) was significantly lower than that of patients receiving long-term ADT (92.8%) (P<0.01). Furthermore, the incidence of cardiovascular toxicity at grade II or above was significantly higher amongst patients treated with short-term ADT compared with patients treated with long-term ADT (P<0.01). Thus, the present study determined that ADT for ≥36 months combined with EBRT significantly improved the progression-free survival of patients with high-risk localized prostate cancer and exhibited an acceptable toxicity profile.
Collapse
Affiliation(s)
- Mitsuru Okubo
- Department of Radiology, Tokyo Medical University Hospital, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Hidetugu Nakayama
- Department of Radiology, Tokyo Medical University Hospital, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Tomohiro Itonaga
- Department of Radiology, Tokyo Medical University Hospital, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Y U Tajima
- Department of Radiology, Tokyo Medical University Hospital, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Sachika Shiraishi
- Department of Radiology, Tokyo Medical University Hospital, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Ryuji Mikami
- Department of Radiology, Tokyo Medical University Hospital, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Akira Sakurada
- Department of Radiology, Tokyo Medical University Hospital, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Shinji Sugahara
- Department of Radiology, Tokyo Medical University Hospital, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Kiyoshi Koizumi
- Department of Radiology, Tokyo Medical University Hospital, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Koichi Tokuuye
- Department of Radiology, Tokyo Medical University Hospital, Shinjuku-ku, Tokyo 160-0023, Japan
| |
Collapse
|
183
|
De Bari B, Vallati M, Gatta R, Simeone C, Girelli G, Ricardi U, Meattini I, Gabriele P, Bellavita R, Krengli M, Cafaro I, Cagna E, Bunkheila F, Borghesi S, Signor M, Di Marco A, Bertoni F, Stefanacci M, Pasinetti N, Buglione M, Magrini SM. Could machine learning improve the prediction of pelvic nodal status of prostate cancer patients? Preliminary results of a pilot study. Cancer Invest 2015; 33:232-40. [PMID: 25950849 DOI: 10.3109/07357907.2015.1024317] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We tested and compared performances of Roach formula, Partin tables and of three Machine Learning (ML) based algorithms based on decision trees in identifying N+ prostate cancer (PC). 1,555 cN0 and 50 cN+ PC were analyzed. Results were also verified on an independent population of 204 operated cN0 patients, with a known pN status (187 pN0, 17 pN1 patients). ML performed better, also when tested on the surgical population, with accuracy, specificity, and sensitivity ranging between 48-86%, 35-91%, and 17-79%, respectively. ML potentially allows better prediction of the nodal status of PC, potentially allowing a better tailoring of pelvic irradiation.
Collapse
Affiliation(s)
- B De Bari
- 1Istituto del Radio "O. Alberti", Radiotherapy Department, Spedali Civili di Brescia and University of Brescia, Brescia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Laramore GE, Zeng J, Fang LMC, Liao JJ, Russell KJ. Pathology Review for Patients with Prostate Cancer Referred to the SCCA Proton Center. Int J Part Ther 2015. [DOI: 10.14338/ijpt-14-00022.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
185
|
Wahba MH, Morad MM. The role of diffusion-weighted MRI: In assessment of response to radiotherapy for prostate cancer. THE EGYPTIAN JOURNAL OF RADIOLOGY AND NUCLEAR MEDICINE 2015. [DOI: 10.1016/j.ejrnm.2014.10.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
186
|
Abstract
The combination of radiation treatment and long-term androgen deprivation therapy (ADT) has been shown in multiple clinical trials to prolong overall survival in men with high-risk prostate cancer compared with either treatment alone. New radiation technologies enable the safe delivery of high radiation doses that improve cancer control compared with lower radiation doses. Based on the results of multiple randomized trials, clinical practice guidelines for high-risk prostate cancer recommend total radiation doses of at least 75.6 Gy, with long-term (2-3 years) ADT. Ongoing research into hypofractionated radiation treatment, whole-pelvic radiation, and combinations of radiation with novel hormonal agents could further improve cancer control and survival outcomes for patients with high-risk prostate cancer.
Collapse
|
187
|
Mason MD, Parulekar WR, Sydes MR, Brundage M, Kirkbride P, Gospodarowicz M, Cowan R, Kostashuk EC, Anderson J, Swanson G, Parmar MKB, Hayter C, Jovic G, Hiltz A, Hetherington J, Sathya J, Barber JBP, McKenzie M, El-Sharkawi S, Souhami L, Hardman PDJ, Chen BE, Warde P. Final Report of the Intergroup Randomized Study of Combined Androgen-Deprivation Therapy Plus Radiotherapy Versus Androgen-Deprivation Therapy Alone in Locally Advanced Prostate Cancer. J Clin Oncol 2015; 33:2143-50. [PMID: 25691677 PMCID: PMC4477786 DOI: 10.1200/jco.2014.57.7510] [Citation(s) in RCA: 184] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Purpose We have previously reported that radiotherapy (RT) added to androgen-deprivation therapy (ADT) improves survival in men with locally advanced prostate cancer. Here, we report the prespecified final analysis of this randomized trial. Patients and Methods NCIC Clinical Trials Group PR.3/Medical Research Council PR07/Intergroup T94-0110 was a randomized controlled trial of patients with locally advanced prostate cancer. Patients with T3-4, N0/Nx, M0 prostate cancer or T1-2 disease with either prostate-specific antigen (PSA) of more than 40 μg/L or PSA of 20 to 40 μg/L plus Gleason score of 8 to 10 were randomly assigned to lifelong ADT alone or to ADT+RT. The RT dose was 64 to 69 Gy in 35 to 39 fractions to the prostate and pelvis or prostate alone. Overall survival was compared using a log-rank test stratified for prespecified variables. Results One thousand two hundred five patients were randomly assigned between 1995 and 2005, 602 to ADT alone and 603 to ADT+RT. At a median follow-up time of 8 years, 465 patients had died, including 199 patients from prostate cancer. Overall survival was significantly improved in the patients allocated to ADT+RT (hazard ratio [HR], 0.70; 95% CI, 0.57 to 0.85; P < .001). Deaths from prostate cancer were significantly reduced by the addition of RT to ADT (HR, 0.46; 95% CI, 0.34 to 0.61; P < .001). Patients on ADT+RT reported a higher frequency of adverse events related to bowel toxicity, but only two of 589 patients had grade 3 or greater diarrhea at 24 months after RT. Conclusion This analysis demonstrates that the previously reported benefit in survival is maintained at a median follow-up of 8 years and firmly establishes the role of RT in the treatment of men with locally advanced prostate cancer.
Collapse
Affiliation(s)
- Malcolm D Mason
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX.
| | - Wendy R Parulekar
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - Matthew R Sydes
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - Michael Brundage
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - Peter Kirkbride
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - Mary Gospodarowicz
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - Richard Cowan
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - Edmund C Kostashuk
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - John Anderson
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - Gregory Swanson
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - Mahesh K B Parmar
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - Charles Hayter
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - Gordana Jovic
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - Andrea Hiltz
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - John Hetherington
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - Jinka Sathya
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - James B P Barber
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - Michael McKenzie
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - Salah El-Sharkawi
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - Luis Souhami
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - P D John Hardman
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - Bingshu E Chen
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| | - Padraig Warde
- Malcolm D. Mason, Cardiff University School of Medicine, Velindre Hospital; James B.P. Barber, Velindre Hospital, Cardiff; Matthew R. Sydes, Mahesh K.B. Parmar, Gordana Jovic, Medical Research Council Clinical Trials Unit at University College London, London; Peter Kirkbride, The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral; Richard Cowan, Christie Hospital, University of Manchester, Manchester; John Anderson, Sheffield Teaching Hospitals, National Health Service Foundation Trust, Sheffield; John Hetherington, Castle Hill Hospital, Hull; Salah El-Sharkawi, South West Wales Cancer Centre, Swansea; P.D. John Hardman, The James Cook University Hospital, Middlesbrough, United Kingdom; Wendy R. Parulekar, Andrea Hiltz, and Bingshu E. Chen, NCIC Clinical Trials Group, Queen's University; Michael Brundage, Cancer Centre of Southeastern Ontario, Kingston; Mary Gospodarowicz and Padraig Warde, University of Toronto, Princess Margaret Cancer Centre; Charles Hayter, University of Toronto, Carlo Fidani Peel Regional Cancer Center, Toronto, Ontario; Edmund C. Kostashuk, Fraser Valley Cancer Centre, Surrey; Michael McKenzie, Vancouver Cancer Centre, Vancouver, British Columbia; Jinka Sathya, Memorial University of Newfoundland, St Johns, Newfoundland and Labrador, Canada; Luis Souhami, McGill University, Montreal, Quebec, Canada; and Gregory Swanson, University of Texas Health Science Center, San Antonio, TX
| |
Collapse
|
188
|
Hata M, Koike I, Wada H, Miyagi E, Kasuya T, Kaizu H, Mukai Y, Inoue T. Postoperative radiation therapy for extramammary Paget's disease. Br J Dermatol 2015; 172:1014-20. [DOI: 10.1111/bjd.13357] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2014] [Indexed: 11/29/2022]
Affiliation(s)
- M. Hata
- Department of Radiology; Yokohama City University Graduate School of Medicine; 3-9 Fukuura Kanazawa-ku 236-0004 Yokohama Kanagawa Japan
| | - I. Koike
- Department of Radiology; Yokohama City University Graduate School of Medicine; 3-9 Fukuura Kanazawa-ku 236-0004 Yokohama Kanagawa Japan
| | - H. Wada
- Department of Dermatology ; Yokohama City University Graduate School of Medicine; 3-9 Fukuura Kanazawa-ku 236-0004 Yokohama Kanagawa Japan
| | - E. Miyagi
- Department of Obstetrics and Gynecology; Yokohama City University Graduate School of Medicine; 3-9 Fukuura Kanazawa-ku 236-0004 Yokohama Kanagawa Japan
| | - T. Kasuya
- Department of Radiology; Yokohama City University Graduate School of Medicine; 3-9 Fukuura Kanazawa-ku 236-0004 Yokohama Kanagawa Japan
| | - H. Kaizu
- Department of Radiology; Yokohama City University Graduate School of Medicine; 3-9 Fukuura Kanazawa-ku 236-0004 Yokohama Kanagawa Japan
| | - Y. Mukai
- Department of Radiology; Yokohama City University Graduate School of Medicine; 3-9 Fukuura Kanazawa-ku 236-0004 Yokohama Kanagawa Japan
| | - T. Inoue
- Department of Radiology; Yokohama City University Graduate School of Medicine; 3-9 Fukuura Kanazawa-ku 236-0004 Yokohama Kanagawa Japan
| |
Collapse
|
189
|
Jani AB, Rossi PJ. Role of radiation and androgen deprivation therapy for advanced prostate cancer. Curr Probl Cancer 2015; 39:41-7. [DOI: 10.1016/j.currproblcancer.2014.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
190
|
Ishii K, Ogino R, Hosokawa Y, Fujioka C, Okada W, Nakahara R, Kawamorita R, Tada T, Hayashi Y, Nakajima T. Whole-pelvic volumetric-modulated arc therapy for high-risk prostate cancer: treatment planning and acute toxicity. JOURNAL OF RADIATION RESEARCH 2015; 56:141-150. [PMID: 25304328 PMCID: PMC4572588 DOI: 10.1093/jrr/rru086] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 08/21/2014] [Accepted: 08/27/2014] [Indexed: 06/04/2023]
Abstract
The objectives of this study were to evaluate dosimetric quality and acute toxicity of volumetric-modulated arc therapy (VMAT) and daily image guidance in high-risk prostate cancer patients. A total of 100 consecutive high-risk prostate cancer patients treated with definitive VMAT with prophylactic whole-pelvic radiotherapy (WPRT) were enrolled. All patients were treated with a double-arc VMAT plan delivering 52 Gy to the prostate planning target volume (PTV), while simultaneously delivering 46.8 Gy to the pelvic nodal PTV in 26 fractions, followed by a single-arc VMAT plan delivering 26 Gy to the prostate PTV in 13 fractions. Image-guided RT was performed with daily cone-beam computed tomography. Dose-volume parameters for the PTV and the organs at risk (OARs), total number of monitor units (MUs) and treatment time were evaluated. Acute toxicity was assessed using the Common Terminology Criteria for Adverse Events, version 4.0. All dosimetric parameters met the present plan acceptance criteria. Mean MU and treatment time were 471 and 146 s for double-arc VMAT, respectively, and were 520 and 76 s for single-arc VMAT, respectively. No Grade 3 or higher acute toxicity was reported. Acute Grade 2 proctitis, diarrhea, and genitourinary toxicity occurred in 12 patients (12%), 6 patients (6%) and 13 patients (13%), respectively. The present study demonstrated that VMAT for WPRT in prostate cancer results in favorable PTV coverage and OAR sparing with short treatment time and an acceptable rate of acute toxicity. These findings support the use of VMAT for delivering WPRT to high-risk prostate cancer patients.
Collapse
Affiliation(s)
- Kentaro Ishii
- Department of Radiation Oncology, Tane General Hospital, 1-12-21 Kujo-minami, Nishi-ku, Osaka, 550-0025, Japan
| | - Ryo Ogino
- Department of Radiation Oncology, Tane General Hospital, 1-12-21 Kujo-minami, Nishi-ku, Osaka, 550-0025, Japan
| | - Yukinari Hosokawa
- Department of Urology, Tane General Hospital, 1-12-21 Kujo-minami, Nishi-ku, Osaka, 550-0025, Japan
| | - Chiaki Fujioka
- Department of Radiation Oncology, Tane General Hospital, 1-12-21 Kujo-minami, Nishi-ku, Osaka, 550-0025, Japan
| | - Wataru Okada
- Department of Radiation Oncology, Tane General Hospital, 1-12-21 Kujo-minami, Nishi-ku, Osaka, 550-0025, Japan
| | - Ryota Nakahara
- Department of Radiation Oncology, Tane General Hospital, 1-12-21 Kujo-minami, Nishi-ku, Osaka, 550-0025, Japan
| | - Ryu Kawamorita
- Department of Radiation Oncology, Tane General Hospital, 1-12-21 Kujo-minami, Nishi-ku, Osaka, 550-0025, Japan
| | - Takuhito Tada
- Department of Radiology, Izumi Municipal Hospital, 4-10-10 Futyu-cho, Izumi, 594-0071, Japan
| | - Yoshiki Hayashi
- Department of Urology, Tane General Hospital, 1-12-21 Kujo-minami, Nishi-ku, Osaka, 550-0025, Japan
| | - Toshifumi Nakajima
- Department of Radiation Oncology, Tane General Hospital, 1-12-21 Kujo-minami, Nishi-ku, Osaka, 550-0025, Japan
| |
Collapse
|
191
|
Weller MA, Kupelian PA, Reddy CA, Stephans KL, Tendulkar RD. Adjuvant versus neoadjuvant androgen deprivation with radiotherapy for prostate cancer: does sequencing matter? Clin Genitourin Cancer 2014; 13:e183-9. [PMID: 25660127 DOI: 10.1016/j.clgc.2014.12.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 12/12/2014] [Accepted: 12/22/2014] [Indexed: 10/24/2022]
Abstract
INTRODUCTION/BACKGROUND Androgen deprivation therapy (ADT) is typically provided neoadjuvantly and concurrently with radiotherapy (RT) in the management of intermediate and high-risk prostate cancer. Our objective was to compare outcomes between patients who received adjuvant ADT (ADJ), ie, immediately after the completion of RT, to those who received a neoadjuvant and concurrent regimen (NEO). MATERIALS AND METHODS From 1995 to 2002, 515 patients with prostate cancer were definitively treated with RT and ADT. NEO was provided 2 to 3 months before the start of RT (n = 311). ADJ was initiated immediately after the completion of RT (n = 204). Kaplan-Meier analysis was used to calculate biochemical relapse-free survival (bRFS), distant metastasis-free survival (DMFS), and overall survival (OS). Cox proportional hazards regression was used to examine the impact of ADT timing on outcomes. RESULTS Ten-year bRFS, DMFS, and OS rates were 61%, 80%, and 66%, respectively. Ten-year bRFS rates for ADJ versus NEO were 63% versus 60% (P = .98). Ten-year DMFS rates for ADJ versus NEO were both 80% (P = .60). Ten-year OS rates for ADJ versus NEO were 65% versus 67% (P = .98). CONCLUSION There was no significant difference in bRFS, DMFS, or OS between neoadjuvant versus adjuvant ADT in the setting of dose-escalated RT for localized prostate cancer. This suggests that the synergy between RT and androgen deprivation is independent of the sequencing of both modalities and that the initiation of RT does not need to be delayed for a course of neoadjuvant ADT.
Collapse
|
192
|
Eom KY, Ha SW, Lee E, Kwak C, Lee SE. Is neoadjuvant androgen deprivation therapy beneficial in prostate cancer treated with definitive radiotherapy? Radiat Oncol J 2014; 32:247-55. [PMID: 25568853 PMCID: PMC4282999 DOI: 10.3857/roj.2014.32.4.247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Revised: 05/31/2014] [Accepted: 12/09/2014] [Indexed: 11/03/2022] Open
Abstract
PURPOSE To determine whether neoadjuvant androgen deprivation therapy (NADT) improves clinical outcomes in patients with prostate cancer treated with definitive radiotherapy. MATERIALS AND METHODS We retrospectively reviewed medical records of 201 patients with prostate cancer treated with radiotherapy between January 1991 and December 2008. Of these, 156 patients with more than 3 years of follow-up were the subjects of this study. The median duration of follow-up was 91.2 months. NADT was given in 103 patients (66%) with median duration of 3.3 months (range, 1.0 to 7.7 months). Radiation dose was escalated gradually from 64 Gy to 81 Gy using intensity-modulated radiotherapy technique. RESULTS Biochemical relapse-free survival (BCRFS) and overall survival (OS) of all patients were 72.6% and 90.7% at 5 years, respectively. BCRFS and OS of NADT group were 79.5% and 89.8% at 5 years and those of radiotherapy alone group were 58.8% and 92.3% at 5 years, respectively. Risk group (p = 0.010) and radiation dose ≥70 Gy (p = 0.017) affected BCRFS independently. NADT was a significant prognostic factor in univariate analysis, but not in multivariate analysis (p = 0.073). Radiation dose ≥70 Gy was only an independent factor for OS (p = 0.007; hazard ratio, 0.261; 95% confidence interval, 0.071-0.963). CONCLUSION NADT prior to definitive radiotherapy did not result in significant benefit in terms of BCRFS and OS. NADT should not be performed routinely in the era of dose-escalated radiotherapy.
Collapse
Affiliation(s)
- Keun-Yong Eom
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea
| | - Sung W Ha
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea. ; Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, Korea. ; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Eunsik Lee
- Department of Urology, Seoul National University College of Medicine, Seoul, Korea
| | - Cheol Kwak
- Department of Urology, Seoul National University College of Medicine, Seoul, Korea
| | - Sang Eun Lee
- Department of Urology, Seoul National University Bundang Hospital, Seongam, Korea
| |
Collapse
|
193
|
Pisansky TM, Hunt D, Gomella LG, Amin MB, Balogh AG, Chinn DM, Seider MJ, Duclos M, Rosenthal SA, Bauman GS, Gore EM, Rotman MZ, Lukka HR, Shipley WU, Dignam JJ, Sandler HM. Duration of androgen suppression before radiotherapy for localized prostate cancer: radiation therapy oncology group randomized clinical trial 9910. J Clin Oncol 2014; 33:332-9. [PMID: 25534388 DOI: 10.1200/jco.2014.58.0662] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To determine whether prolonged androgen suppression (AS) duration before radiotherapy improves survival and disease control in prostate cancer. PATIENTS AND METHODS One thousand five hundred seventy-nine men with intermediate-risk prostate cancer were randomly assigned to 8 weeks of AS followed by radiotherapy with an additional 8 weeks of concurrent AS (16 weeks total) or to 28 weeks of AS followed by radiotherapy with an additional 8 weeks of AS (36 weeks total). The trial sought primarily to detect a 33% reduction in the hazard of prostate cancer death in the 28-week assignment. Time-to-event end points are reported for up to 10 years of follow-up. RESULTS There were no between-group differences in baseline characteristics of 1,489 eligible patients with follow-up. For the 8- and 28-week assignments, 10-year disease-specific survival rates were 95% (95% CI, 93.3% to 97.0%) and 96% (95% CI, 94.6% to 98.0%; hazard ratio [HR], 0.81; P = .45), respectively, and 10-year overall survival rates were 66% (95% CI, 62.0% to 69.9%) and 67% (95% CI, 63.0% to 70.8%; HR, 0.95; P = .62), respectively. For the 8- and 28-week assignments, 10-year cumulative incidences of locoregional progression were 6% (95% CI, 4.3% to 8.0%) and 4% (95% CI, 2.5% to 5.7%; HR, 0.65; P = .07), respectively; 10-year distant metastasis cumulative incidences were 6% (95% CI, 4.0% to 7.7%) and 6% (95% CI, 4.0% to 7.6%; HR, 1.07; P = .80), respectively; and 10-year prostate-specific antigen-based recurrence cumulative incidences were 27% (95% CI, 23.1% to 29.8%) and 27% (95% CI, 23.4% to 30.3%; HR, 0.97; P = .77), respectively. CONCLUSION Extending AS duration from 8 weeks to 28 weeks before radiotherapy did not improve outcomes. A lower than expected prostate cancer death rate reduced ability to detect a between-group difference in disease-specific survival. The schedule of 8 weeks of AS before radiotherapy plus 8 weeks of AS during radiotherapy remains a standard of care in intermediate-risk prostate cancer.
Collapse
Affiliation(s)
- Thomas M Pisansky
- Thomas M. Pisansky, Mayo Clinic, Rochester, MN; Daniel Hunt and James J. Dignam, Radiation Therapy Oncology Group Statistical Center; Leonard G. Gomella, Thomas Jefferson University, Philadelphia, PA; Mahul B. Amin and Howard M. Sandler, Cedars-Sinai Medical Center, Los Angeles; Daniel M. Chinn, John Muir Medical Center-Concord Campus, Concord; Seth A. Rosenthal, Sutter Medical Group, Sacramento, CA; Michael J. Seider, Akron City Hospital, Akron, OH; Elizabeth M. Gore, Zablocki Veterans Administration Medical Center-Wood, Milwaukee, WI; Marvin Z. Rotman, Brooklyn Minority-Based Community Clinical Oncology Program, State University of New York Downstate, Brooklyn, NY; William U. Shipley, Massachusetts General Hospital, Boston, MA; James J. Dignam, University of Chicago, Chicago, IL; Alexander G. Balogh, Tom Baker Cancer Centre, Calgary, Alberta; Marie Duclos, McGill University, Montreal, Quebec; Glenn S. Bauman, London Regional Cancer Program, London; and Himanshu R. Lukka, McMaster University, Hamilton, Ontario, Canada.
| | - Daniel Hunt
- Thomas M. Pisansky, Mayo Clinic, Rochester, MN; Daniel Hunt and James J. Dignam, Radiation Therapy Oncology Group Statistical Center; Leonard G. Gomella, Thomas Jefferson University, Philadelphia, PA; Mahul B. Amin and Howard M. Sandler, Cedars-Sinai Medical Center, Los Angeles; Daniel M. Chinn, John Muir Medical Center-Concord Campus, Concord; Seth A. Rosenthal, Sutter Medical Group, Sacramento, CA; Michael J. Seider, Akron City Hospital, Akron, OH; Elizabeth M. Gore, Zablocki Veterans Administration Medical Center-Wood, Milwaukee, WI; Marvin Z. Rotman, Brooklyn Minority-Based Community Clinical Oncology Program, State University of New York Downstate, Brooklyn, NY; William U. Shipley, Massachusetts General Hospital, Boston, MA; James J. Dignam, University of Chicago, Chicago, IL; Alexander G. Balogh, Tom Baker Cancer Centre, Calgary, Alberta; Marie Duclos, McGill University, Montreal, Quebec; Glenn S. Bauman, London Regional Cancer Program, London; and Himanshu R. Lukka, McMaster University, Hamilton, Ontario, Canada
| | - Leonard G Gomella
- Thomas M. Pisansky, Mayo Clinic, Rochester, MN; Daniel Hunt and James J. Dignam, Radiation Therapy Oncology Group Statistical Center; Leonard G. Gomella, Thomas Jefferson University, Philadelphia, PA; Mahul B. Amin and Howard M. Sandler, Cedars-Sinai Medical Center, Los Angeles; Daniel M. Chinn, John Muir Medical Center-Concord Campus, Concord; Seth A. Rosenthal, Sutter Medical Group, Sacramento, CA; Michael J. Seider, Akron City Hospital, Akron, OH; Elizabeth M. Gore, Zablocki Veterans Administration Medical Center-Wood, Milwaukee, WI; Marvin Z. Rotman, Brooklyn Minority-Based Community Clinical Oncology Program, State University of New York Downstate, Brooklyn, NY; William U. Shipley, Massachusetts General Hospital, Boston, MA; James J. Dignam, University of Chicago, Chicago, IL; Alexander G. Balogh, Tom Baker Cancer Centre, Calgary, Alberta; Marie Duclos, McGill University, Montreal, Quebec; Glenn S. Bauman, London Regional Cancer Program, London; and Himanshu R. Lukka, McMaster University, Hamilton, Ontario, Canada
| | - Mahul B Amin
- Thomas M. Pisansky, Mayo Clinic, Rochester, MN; Daniel Hunt and James J. Dignam, Radiation Therapy Oncology Group Statistical Center; Leonard G. Gomella, Thomas Jefferson University, Philadelphia, PA; Mahul B. Amin and Howard M. Sandler, Cedars-Sinai Medical Center, Los Angeles; Daniel M. Chinn, John Muir Medical Center-Concord Campus, Concord; Seth A. Rosenthal, Sutter Medical Group, Sacramento, CA; Michael J. Seider, Akron City Hospital, Akron, OH; Elizabeth M. Gore, Zablocki Veterans Administration Medical Center-Wood, Milwaukee, WI; Marvin Z. Rotman, Brooklyn Minority-Based Community Clinical Oncology Program, State University of New York Downstate, Brooklyn, NY; William U. Shipley, Massachusetts General Hospital, Boston, MA; James J. Dignam, University of Chicago, Chicago, IL; Alexander G. Balogh, Tom Baker Cancer Centre, Calgary, Alberta; Marie Duclos, McGill University, Montreal, Quebec; Glenn S. Bauman, London Regional Cancer Program, London; and Himanshu R. Lukka, McMaster University, Hamilton, Ontario, Canada
| | - Alexander G Balogh
- Thomas M. Pisansky, Mayo Clinic, Rochester, MN; Daniel Hunt and James J. Dignam, Radiation Therapy Oncology Group Statistical Center; Leonard G. Gomella, Thomas Jefferson University, Philadelphia, PA; Mahul B. Amin and Howard M. Sandler, Cedars-Sinai Medical Center, Los Angeles; Daniel M. Chinn, John Muir Medical Center-Concord Campus, Concord; Seth A. Rosenthal, Sutter Medical Group, Sacramento, CA; Michael J. Seider, Akron City Hospital, Akron, OH; Elizabeth M. Gore, Zablocki Veterans Administration Medical Center-Wood, Milwaukee, WI; Marvin Z. Rotman, Brooklyn Minority-Based Community Clinical Oncology Program, State University of New York Downstate, Brooklyn, NY; William U. Shipley, Massachusetts General Hospital, Boston, MA; James J. Dignam, University of Chicago, Chicago, IL; Alexander G. Balogh, Tom Baker Cancer Centre, Calgary, Alberta; Marie Duclos, McGill University, Montreal, Quebec; Glenn S. Bauman, London Regional Cancer Program, London; and Himanshu R. Lukka, McMaster University, Hamilton, Ontario, Canada
| | - Daniel M Chinn
- Thomas M. Pisansky, Mayo Clinic, Rochester, MN; Daniel Hunt and James J. Dignam, Radiation Therapy Oncology Group Statistical Center; Leonard G. Gomella, Thomas Jefferson University, Philadelphia, PA; Mahul B. Amin and Howard M. Sandler, Cedars-Sinai Medical Center, Los Angeles; Daniel M. Chinn, John Muir Medical Center-Concord Campus, Concord; Seth A. Rosenthal, Sutter Medical Group, Sacramento, CA; Michael J. Seider, Akron City Hospital, Akron, OH; Elizabeth M. Gore, Zablocki Veterans Administration Medical Center-Wood, Milwaukee, WI; Marvin Z. Rotman, Brooklyn Minority-Based Community Clinical Oncology Program, State University of New York Downstate, Brooklyn, NY; William U. Shipley, Massachusetts General Hospital, Boston, MA; James J. Dignam, University of Chicago, Chicago, IL; Alexander G. Balogh, Tom Baker Cancer Centre, Calgary, Alberta; Marie Duclos, McGill University, Montreal, Quebec; Glenn S. Bauman, London Regional Cancer Program, London; and Himanshu R. Lukka, McMaster University, Hamilton, Ontario, Canada
| | - Michael J Seider
- Thomas M. Pisansky, Mayo Clinic, Rochester, MN; Daniel Hunt and James J. Dignam, Radiation Therapy Oncology Group Statistical Center; Leonard G. Gomella, Thomas Jefferson University, Philadelphia, PA; Mahul B. Amin and Howard M. Sandler, Cedars-Sinai Medical Center, Los Angeles; Daniel M. Chinn, John Muir Medical Center-Concord Campus, Concord; Seth A. Rosenthal, Sutter Medical Group, Sacramento, CA; Michael J. Seider, Akron City Hospital, Akron, OH; Elizabeth M. Gore, Zablocki Veterans Administration Medical Center-Wood, Milwaukee, WI; Marvin Z. Rotman, Brooklyn Minority-Based Community Clinical Oncology Program, State University of New York Downstate, Brooklyn, NY; William U. Shipley, Massachusetts General Hospital, Boston, MA; James J. Dignam, University of Chicago, Chicago, IL; Alexander G. Balogh, Tom Baker Cancer Centre, Calgary, Alberta; Marie Duclos, McGill University, Montreal, Quebec; Glenn S. Bauman, London Regional Cancer Program, London; and Himanshu R. Lukka, McMaster University, Hamilton, Ontario, Canada
| | - Marie Duclos
- Thomas M. Pisansky, Mayo Clinic, Rochester, MN; Daniel Hunt and James J. Dignam, Radiation Therapy Oncology Group Statistical Center; Leonard G. Gomella, Thomas Jefferson University, Philadelphia, PA; Mahul B. Amin and Howard M. Sandler, Cedars-Sinai Medical Center, Los Angeles; Daniel M. Chinn, John Muir Medical Center-Concord Campus, Concord; Seth A. Rosenthal, Sutter Medical Group, Sacramento, CA; Michael J. Seider, Akron City Hospital, Akron, OH; Elizabeth M. Gore, Zablocki Veterans Administration Medical Center-Wood, Milwaukee, WI; Marvin Z. Rotman, Brooklyn Minority-Based Community Clinical Oncology Program, State University of New York Downstate, Brooklyn, NY; William U. Shipley, Massachusetts General Hospital, Boston, MA; James J. Dignam, University of Chicago, Chicago, IL; Alexander G. Balogh, Tom Baker Cancer Centre, Calgary, Alberta; Marie Duclos, McGill University, Montreal, Quebec; Glenn S. Bauman, London Regional Cancer Program, London; and Himanshu R. Lukka, McMaster University, Hamilton, Ontario, Canada
| | - Seth A Rosenthal
- Thomas M. Pisansky, Mayo Clinic, Rochester, MN; Daniel Hunt and James J. Dignam, Radiation Therapy Oncology Group Statistical Center; Leonard G. Gomella, Thomas Jefferson University, Philadelphia, PA; Mahul B. Amin and Howard M. Sandler, Cedars-Sinai Medical Center, Los Angeles; Daniel M. Chinn, John Muir Medical Center-Concord Campus, Concord; Seth A. Rosenthal, Sutter Medical Group, Sacramento, CA; Michael J. Seider, Akron City Hospital, Akron, OH; Elizabeth M. Gore, Zablocki Veterans Administration Medical Center-Wood, Milwaukee, WI; Marvin Z. Rotman, Brooklyn Minority-Based Community Clinical Oncology Program, State University of New York Downstate, Brooklyn, NY; William U. Shipley, Massachusetts General Hospital, Boston, MA; James J. Dignam, University of Chicago, Chicago, IL; Alexander G. Balogh, Tom Baker Cancer Centre, Calgary, Alberta; Marie Duclos, McGill University, Montreal, Quebec; Glenn S. Bauman, London Regional Cancer Program, London; and Himanshu R. Lukka, McMaster University, Hamilton, Ontario, Canada
| | - Glenn S Bauman
- Thomas M. Pisansky, Mayo Clinic, Rochester, MN; Daniel Hunt and James J. Dignam, Radiation Therapy Oncology Group Statistical Center; Leonard G. Gomella, Thomas Jefferson University, Philadelphia, PA; Mahul B. Amin and Howard M. Sandler, Cedars-Sinai Medical Center, Los Angeles; Daniel M. Chinn, John Muir Medical Center-Concord Campus, Concord; Seth A. Rosenthal, Sutter Medical Group, Sacramento, CA; Michael J. Seider, Akron City Hospital, Akron, OH; Elizabeth M. Gore, Zablocki Veterans Administration Medical Center-Wood, Milwaukee, WI; Marvin Z. Rotman, Brooklyn Minority-Based Community Clinical Oncology Program, State University of New York Downstate, Brooklyn, NY; William U. Shipley, Massachusetts General Hospital, Boston, MA; James J. Dignam, University of Chicago, Chicago, IL; Alexander G. Balogh, Tom Baker Cancer Centre, Calgary, Alberta; Marie Duclos, McGill University, Montreal, Quebec; Glenn S. Bauman, London Regional Cancer Program, London; and Himanshu R. Lukka, McMaster University, Hamilton, Ontario, Canada
| | - Elizabeth M Gore
- Thomas M. Pisansky, Mayo Clinic, Rochester, MN; Daniel Hunt and James J. Dignam, Radiation Therapy Oncology Group Statistical Center; Leonard G. Gomella, Thomas Jefferson University, Philadelphia, PA; Mahul B. Amin and Howard M. Sandler, Cedars-Sinai Medical Center, Los Angeles; Daniel M. Chinn, John Muir Medical Center-Concord Campus, Concord; Seth A. Rosenthal, Sutter Medical Group, Sacramento, CA; Michael J. Seider, Akron City Hospital, Akron, OH; Elizabeth M. Gore, Zablocki Veterans Administration Medical Center-Wood, Milwaukee, WI; Marvin Z. Rotman, Brooklyn Minority-Based Community Clinical Oncology Program, State University of New York Downstate, Brooklyn, NY; William U. Shipley, Massachusetts General Hospital, Boston, MA; James J. Dignam, University of Chicago, Chicago, IL; Alexander G. Balogh, Tom Baker Cancer Centre, Calgary, Alberta; Marie Duclos, McGill University, Montreal, Quebec; Glenn S. Bauman, London Regional Cancer Program, London; and Himanshu R. Lukka, McMaster University, Hamilton, Ontario, Canada
| | - Marvin Z Rotman
- Thomas M. Pisansky, Mayo Clinic, Rochester, MN; Daniel Hunt and James J. Dignam, Radiation Therapy Oncology Group Statistical Center; Leonard G. Gomella, Thomas Jefferson University, Philadelphia, PA; Mahul B. Amin and Howard M. Sandler, Cedars-Sinai Medical Center, Los Angeles; Daniel M. Chinn, John Muir Medical Center-Concord Campus, Concord; Seth A. Rosenthal, Sutter Medical Group, Sacramento, CA; Michael J. Seider, Akron City Hospital, Akron, OH; Elizabeth M. Gore, Zablocki Veterans Administration Medical Center-Wood, Milwaukee, WI; Marvin Z. Rotman, Brooklyn Minority-Based Community Clinical Oncology Program, State University of New York Downstate, Brooklyn, NY; William U. Shipley, Massachusetts General Hospital, Boston, MA; James J. Dignam, University of Chicago, Chicago, IL; Alexander G. Balogh, Tom Baker Cancer Centre, Calgary, Alberta; Marie Duclos, McGill University, Montreal, Quebec; Glenn S. Bauman, London Regional Cancer Program, London; and Himanshu R. Lukka, McMaster University, Hamilton, Ontario, Canada
| | - Himanshu R Lukka
- Thomas M. Pisansky, Mayo Clinic, Rochester, MN; Daniel Hunt and James J. Dignam, Radiation Therapy Oncology Group Statistical Center; Leonard G. Gomella, Thomas Jefferson University, Philadelphia, PA; Mahul B. Amin and Howard M. Sandler, Cedars-Sinai Medical Center, Los Angeles; Daniel M. Chinn, John Muir Medical Center-Concord Campus, Concord; Seth A. Rosenthal, Sutter Medical Group, Sacramento, CA; Michael J. Seider, Akron City Hospital, Akron, OH; Elizabeth M. Gore, Zablocki Veterans Administration Medical Center-Wood, Milwaukee, WI; Marvin Z. Rotman, Brooklyn Minority-Based Community Clinical Oncology Program, State University of New York Downstate, Brooklyn, NY; William U. Shipley, Massachusetts General Hospital, Boston, MA; James J. Dignam, University of Chicago, Chicago, IL; Alexander G. Balogh, Tom Baker Cancer Centre, Calgary, Alberta; Marie Duclos, McGill University, Montreal, Quebec; Glenn S. Bauman, London Regional Cancer Program, London; and Himanshu R. Lukka, McMaster University, Hamilton, Ontario, Canada
| | - William U Shipley
- Thomas M. Pisansky, Mayo Clinic, Rochester, MN; Daniel Hunt and James J. Dignam, Radiation Therapy Oncology Group Statistical Center; Leonard G. Gomella, Thomas Jefferson University, Philadelphia, PA; Mahul B. Amin and Howard M. Sandler, Cedars-Sinai Medical Center, Los Angeles; Daniel M. Chinn, John Muir Medical Center-Concord Campus, Concord; Seth A. Rosenthal, Sutter Medical Group, Sacramento, CA; Michael J. Seider, Akron City Hospital, Akron, OH; Elizabeth M. Gore, Zablocki Veterans Administration Medical Center-Wood, Milwaukee, WI; Marvin Z. Rotman, Brooklyn Minority-Based Community Clinical Oncology Program, State University of New York Downstate, Brooklyn, NY; William U. Shipley, Massachusetts General Hospital, Boston, MA; James J. Dignam, University of Chicago, Chicago, IL; Alexander G. Balogh, Tom Baker Cancer Centre, Calgary, Alberta; Marie Duclos, McGill University, Montreal, Quebec; Glenn S. Bauman, London Regional Cancer Program, London; and Himanshu R. Lukka, McMaster University, Hamilton, Ontario, Canada
| | - James J Dignam
- Thomas M. Pisansky, Mayo Clinic, Rochester, MN; Daniel Hunt and James J. Dignam, Radiation Therapy Oncology Group Statistical Center; Leonard G. Gomella, Thomas Jefferson University, Philadelphia, PA; Mahul B. Amin and Howard M. Sandler, Cedars-Sinai Medical Center, Los Angeles; Daniel M. Chinn, John Muir Medical Center-Concord Campus, Concord; Seth A. Rosenthal, Sutter Medical Group, Sacramento, CA; Michael J. Seider, Akron City Hospital, Akron, OH; Elizabeth M. Gore, Zablocki Veterans Administration Medical Center-Wood, Milwaukee, WI; Marvin Z. Rotman, Brooklyn Minority-Based Community Clinical Oncology Program, State University of New York Downstate, Brooklyn, NY; William U. Shipley, Massachusetts General Hospital, Boston, MA; James J. Dignam, University of Chicago, Chicago, IL; Alexander G. Balogh, Tom Baker Cancer Centre, Calgary, Alberta; Marie Duclos, McGill University, Montreal, Quebec; Glenn S. Bauman, London Regional Cancer Program, London; and Himanshu R. Lukka, McMaster University, Hamilton, Ontario, Canada
| | - Howard M Sandler
- Thomas M. Pisansky, Mayo Clinic, Rochester, MN; Daniel Hunt and James J. Dignam, Radiation Therapy Oncology Group Statistical Center; Leonard G. Gomella, Thomas Jefferson University, Philadelphia, PA; Mahul B. Amin and Howard M. Sandler, Cedars-Sinai Medical Center, Los Angeles; Daniel M. Chinn, John Muir Medical Center-Concord Campus, Concord; Seth A. Rosenthal, Sutter Medical Group, Sacramento, CA; Michael J. Seider, Akron City Hospital, Akron, OH; Elizabeth M. Gore, Zablocki Veterans Administration Medical Center-Wood, Milwaukee, WI; Marvin Z. Rotman, Brooklyn Minority-Based Community Clinical Oncology Program, State University of New York Downstate, Brooklyn, NY; William U. Shipley, Massachusetts General Hospital, Boston, MA; James J. Dignam, University of Chicago, Chicago, IL; Alexander G. Balogh, Tom Baker Cancer Centre, Calgary, Alberta; Marie Duclos, McGill University, Montreal, Quebec; Glenn S. Bauman, London Regional Cancer Program, London; and Himanshu R. Lukka, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
194
|
Pinkawa M, Djukic V, Klotz J, Petz D, Piroth MD, Holy R, Eble MJ. Hematologic changes during prostate cancer radiation therapy are dependent on the treatment volume. Future Oncol 2014; 10:835-43. [PMID: 24799064 DOI: 10.2217/fon.13.237] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To assess hematologic changes of modern prostate radiation therapy (RT) comparing different target volumes. PATIENTS & METHODS Blood samples were evaluated before (T1), during (T2-T4) and 6-8 weeks after (T5) RT in a group of 113 patients. Whole-pelvic RT up to 46 Gy was applied in 27 cases. The total dose to the prostatic fossa (n = 46)/prostate (n = 67) was 66/76 Gy. RESULTS Erythrocyte, leukocyte and platelet levels decreased significantly relative to baseline levels at T2-T5. Neoadjuvant hormonal therapy had an impact on hemoglobin levels before and during RT. The cumulative incidence of grade 2 leukopenia was 15 versus 2% (p = 0.02) and grade 2 anemia 8 versus 0% (p = 0.03) with versus without whole-pelvic RT, respectively. Lymphocyte decrease was larger at times T2-T5 (36 vs 3% grade 3 toxicity; p < 0.01). CONCLUSION Prostate RT has a small but significant and longer effect on the blood count. Lower lymphocyte levels need to be considered when larger volumes are treated.
Collapse
Affiliation(s)
- Michael Pinkawa
- Department of Radiation Oncology, Rheinisch-Westfaelische Technische Hochschule Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany
| | | | | | | | | | | | | |
Collapse
|
195
|
Musunuru HB, Cheung P, Loblaw A. Evolution of hypofractionated accelerated radiotherapy for prostate cancer - the sunnybrook experience. Front Oncol 2014; 4:313. [PMID: 25452934 PMCID: PMC4231839 DOI: 10.3389/fonc.2014.00313] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 10/20/2014] [Indexed: 11/13/2022] Open
Abstract
Stereotactic ablative body radiotherapy (SABR) is a newer method of ultra hypo fractionated radiotherapy that uses combination of image-guided radiotherapy (IGRT) and intensity-modulated radiotherapy (IMRT) or volumetric modulated arc therapy (VMAT), to deliver high doses of radiation in a few fractions to a target, at the same time sparing the surrounding organs at risk (OAR). SABR is ideal for treating small volumes of disease and has been introduced in a number of disease sites including brain, lung, liver, spine, and prostate. Given the radiobiological advantages of treating prostate cancer with high doses per fraction, SABR is becoming a standard of care for low and intermediate-risk prostate cancer patients based upon the results from Sunnybrook and also the US-based prostate SABR consortium. This review examines the development of moderate and ultra hypo-fractionation schedules at the Odette Cancer centre, Sunnybrook Health Sciences. Moderate hypo-fractionation protocol was first developed in 2001 for intermediate-risk prostate cancer and from there on different treatment schedules including SABR evolved for all risk groups.
Collapse
Affiliation(s)
- Hima Bindu Musunuru
- Odette Cancer Centre, Sunnybrook Health Sciences Centre , Toronto, ON , Canada ; Department of Radiation Oncology, University of Toronto , Toronto, ON , Canada
| | - Patrick Cheung
- Odette Cancer Centre, Sunnybrook Health Sciences Centre , Toronto, ON , Canada ; Department of Radiation Oncology, University of Toronto , Toronto, ON , Canada
| | - Andrew Loblaw
- Odette Cancer Centre, Sunnybrook Health Sciences Centre , Toronto, ON , Canada ; Department of Radiation Oncology, University of Toronto , Toronto, ON , Canada ; Department of Health Policy, Measurement and Evaluation, University of Toronto , Toronto, ON , Canada
| |
Collapse
|
196
|
Martin NE, D'Amico AV. Progress and controversies: Radiation therapy for prostate cancer. CA Cancer J Clin 2014; 64:389-407. [PMID: 25234700 DOI: 10.3322/caac.21250] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 08/14/2014] [Accepted: 08/15/2014] [Indexed: 12/14/2022] Open
Abstract
Radiation therapy remains a standard treatment option for men with localized prostate cancer. Alone or in combination with androgen-deprivation therapy, it represents a curative treatment and has been shown to prolong survival in selected populations. In this article, the authors review recent advances in prostate radiation-treatment techniques, photon versus proton radiation, modification of treatment fractionation, and brachytherapy-all focusing on disease control and the impact on morbidity. Also discussed are refinements in the risk stratification of men with prostate cancer and how these are better for matching patients to appropriate treatment, particularly around combined androgen-deprivation therapy. Many of these advances have cost and treatment burden implications, which have significant repercussions given the prevalence of prostate cancer. The discussion includes approaches to improve value and future directions for research.
Collapse
Affiliation(s)
- Neil E Martin
- Assistant Professor, Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | | |
Collapse
|
197
|
Lin YW, Lin LC, Lin KL. The early result of whole pelvic radiotherapy and stereotactic body radiotherapy boost for high-risk localized prostate cancer. Front Oncol 2014; 4:278. [PMID: 25401085 PMCID: PMC4215618 DOI: 10.3389/fonc.2014.00278] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 09/26/2014] [Indexed: 11/13/2022] Open
Abstract
Purpose: The rationale for hypofractionated radiotherapy in the treatment of prostate cancer is based on the modern understanding of radiobiology and advances in stereotactic body radiotherapy (SBRT) techniques. Whole-pelvis irradiation combined with SBRT boost for high-risk prostate cancer might escalate biologically effective dose without increasing toxicity. Here, we report our 4-year results of SBRT boost for high-risk localized prostate cancer. Methods and Materials: From October 2009 to August 2012, 41 patients newly diagnosed, high-risk or very high-risk (NCCN definition) localized prostate cancer were treated with whole-pelvis irradiation and SBRT boost. The whole pelvis dose was 45 Gy (25 fractions of 1.8 Gy). The SBRT boost dose was 21 Gy (three fractions of 7 Gy). Ninety percent of these patients received hormone therapy. The toxicities of gastrointestinal (GI) and genitourinary (GU) tracts were scored by Common Toxicity Criteria Adverse Effect (CTCAE v3.0). Biochemical failure was defined by Phoenix definition. Results: Median follow-up was 42 months. Mean PSA before treatment was 44.18 ng/ml. Mean PSA level at 3, 6, 12, 18, and 24 months was 0.94, 0.44, 0.13, 0.12, and 0.05 ng/ml, respectively. The estimated 4-year biochemical failure-free survival was 91.9%. Three biochemical failures were observed. GI and GU tract toxicities were minimal. No grade 3 acute GU or GI toxicity was noted. During radiation therapy, 27% of the patient had grade 2 acute GU toxicity and 12% had grade 2 acute GI toxicity. At 3 months, most toxicity scores had returned to baseline. At the last follow-up, there was no grade 3 late GU or GI toxicity. Conclusions: Whole-pelvis irradiation combined with SBRT boost for high-risk localized prostate cancer is feasible with minimal toxicity and encouraging biochemical failure-free survival. Continued accrual and follow-up would be necessary to confirm the biochemical control rate and the toxicity profiles.
Collapse
Affiliation(s)
- Yu-Wei Lin
- Department of Radiation Oncology, Chi Mei Medical Center , Tainan , Taiwan ; Institute of Biomedical Sciences, National Sun Yat-sen University , Kaohsiung , Taiwan ; The School of Medicine, Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Li-Ching Lin
- Department of Radiation Oncology, Chi Mei Medical Center , Tainan , Taiwan ; School of Medicine, Taipei Medical University , Taipei , Taiwan
| | - Kuei-Li Lin
- Department of Radiation Oncology, Chi Mei Medical Center , Tainan , Taiwan
| |
Collapse
|
198
|
Valeriani M, Carnevale A, Osti MF, DE Sanctis V, Agolli L, Maurizi Enrici R. Image guided intensity modulated hypofractionated radiotherapy in high-risk prostate cancer patients treated four or five times per week: analysis of toxicity and preliminary results. Radiat Oncol 2014; 9:214. [PMID: 25260377 PMCID: PMC4261590 DOI: 10.1186/1748-717x-9-214] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 09/15/2014] [Indexed: 11/10/2022] Open
Abstract
Background To evaluate efficacy and toxicity of hypofractionated intensity-modulated simultaneous integrated boost (IMRT-SIB) and image-guided (IGRT) radiotherapy in the treatment of high-risk prostate cancer patients. Methods Eighty-two patients with high-risk prostate cancer were analysed. An IMRT treatment was planned delivering 68.75 Gy to the prostate, 55 Gy to the seminal vesicles and positive nodes and 45 Gy to the pelvis in 25 fractions. The first 59 patients received 4 weekly fractions whereas the last 23 patients received 5 weekly fractions. All patients were submitted to hormonal therapy. Results The median follow-up was 31 months. Acute grade 1–2 gastrointestinal (GI) toxicity rates were 13.4%. Grade 1–2 and grade 3 genitourinary (GU) toxicity rates were 22% and 1.2%, respectively. Grade 1 and 2 GI late toxicity rates were 1.2%. No grade ≥3 toxicity was recorded. Grade 1 GU late toxicity rate was 2.4%. No grade ≥2 toxicity was recorded. No significant difference was calculated in terms of acute and late toxicity between the group treated 4 or 5 times weekly. The actuarial 3-years Overall survival and Freedom from biochemical failure were 98.6% and 91.3%, respectively. Conclusions The present study demonstrated that hypofractionated IGRT-IMRT-SIB in patients with high-risk prostate cancer is efficient with acceptable toxicity profile. Outcome in terms of survival are promising, but longer follow-up is needed.
Collapse
Affiliation(s)
- Maurizio Valeriani
- Department of Radiation Oncology, La Sapienza" University, Sant'Andrea Hospital of Rome, Rome, Italy.
| | | | | | | | | | | |
Collapse
|
199
|
Kim H, Kim JW, Hong SJ, Rha KH, Lee CG, Yang SC, Choi YD, Suh CO, Cho J. Treatment outcome of localized prostate cancer by 70 Gy hypofractionated intensity-modulated radiotherapy with a customized rectal balloon. Radiat Oncol J 2014; 32:187-197. [PMID: 25324991 PMCID: PMC4194302 DOI: 10.3857/roj.2014.32.3.187] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 08/03/2014] [Accepted: 09/01/2014] [Indexed: 12/30/2022] Open
Abstract
PURPOSE We aimed to analyze the treatment outcome and long-term toxicity of 70 Gy hypofractionated intensity-modulated radiotherapy (IMRT) for localized prostate cancer using a customized rectal balloon. MATERIALS AND METHODS We reviewed medical records of 86 prostate cancer patients who received curative radiotherapy between January 2004 and December 2011 at our institution. Patients were designated as low (12.8%), intermediate (20.9%), or high risk (66.3%). Thirty patients received a total dose of 70 Gy in 28 fractions over 5 weeks via IMRT (the Hypo-IMRT group); 56 received 70.2 Gy in 39 fractions over 7 weeks via 3-dimensional conformal radiotherapy (the CF-3DRT group, which served as a reference for comparison). A customized rectal balloon was placed in Hypo-IMRT group throughout the entire radiotherapy course. Androgen deprivation therapy was administered to 47 patients (Hypo-IMRT group, 17; CF-3DRT group, 30). Late genitourinary (GU) and gastrointestinal (GI) toxicity were evaluated according to the Radiation Therapy Oncology Group criteria. RESULTS The median follow-up period was 74.4 months (range, 18.8 to 125.9 months). The 5-year actuarial biochemical relapse-free survival rates for low-, intermediate-, and high-risk patients were 100%, 100%, and 88.5%, respectively, for the Hypo-IMRT group and 80%, 77.8%, and 63.6%, respectively, for the CF-3DRT group (p < 0.046). No patient presented with acute or late GU toxicity ≥grade 3. Late grade 3 GI toxicity occurred in 2 patients (3.6%) in the CF-3DRT group and 1 patient (3.3%) in the Hypo-IMRT group. CONCLUSION Hypo-IMRT with a customized rectal balloon resulted in excellent biochemical control rates with minimal toxicity in localized prostate cancer patients.
Collapse
Affiliation(s)
- Hyunjung Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Jun Won Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Joon Hong
- Department of Urology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Koon Ho Rha
- Department of Urology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Chang-Geol Lee
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Choul Yang
- Department of Urology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Young Deuk Choi
- Department of Urology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Chang-Ok Suh
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Jaeho Cho
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
200
|
Toxicity and quality of life after choline-PET/CT directed salvage lymph node dissection and adjuvant radiotherapy in nodal recurrent prostate cancer. Radiat Oncol 2014; 9:178. [PMID: 25112785 PMCID: PMC4251989 DOI: 10.1186/1748-717x-9-178] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 08/07/2014] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND In a previous study we demonstrated that, based on 11C/18 F-choline positron emission tomography-computerized-tomography as a diagnostic tool, salvage lymph node dissection (LND) plus adjuvant radiotherapy (ART) is feasible for treatment of pelvic/retroperitoneal nodal recurrence of prostate cancer (PCa). However, the toxicity of this combined treatment strategy has not been systematically investigated before. The aim of the current study was to evaluate the acute and late toxicity and quality of life of ART after LND in pelvic/retroperitoneal nodal recurrent PCa. MATERIAL AND METHODS 43 patients with nodal recurrent PCa were treated with 46 LND followed by ART (mean 49.6 Gy total dose) at the sites of nodal recurrence. Toxicity of ART was analysed by physically examination (31/43, 72.1%), by requesting 15 frequent items of adverse events from the Common-Terminology-Criteria for Adverse Events Version 4.0-catalogue and by review of medical records. QLQ-C30 (EORTC quality of life assessment) and PR25 (prostate cancer module) questionnaires were used to investigate quality of life. Toxicity was evaluated before starting of ART, during ART (acute toxicity), after ART (mean 2.3 months) and at end of follow up (mean 3.2 years after end of ART) reflecting late toxicity. RESULTS 71.7% (33/46) of 46 ART were treatment of pelvic, 10.9% (5/46) of retroperitoneal only and 28.3% (13/46) of pelvic and retroperitoneal regions. Overall 52 symptoms representing toxicities were observed before ART, 107 during ART, 88 after end of ART and 52 at latest follow up. Leading toxicities during ART were diarrhoea (19%, 20/107), urinary incontinence (16%, 17/107) and fatigue (16%, 17/107). The spectrum of late toxicities was almost equal to those before beginning of ART. No grade 3 adverse events or chronic lymphedema at extremities were observed. We observed no clear correlation between localisation of treated regions, technique of ART and frequency or severity of toxicities. Mean quality of life at final evaluation was 74%. CONCLUSION ART after extended LND in PCa relapse is justifiable with respect to adverse effects and toxicity. The side effects were circumscribed and well tolerated. The spectrum of adverse events at latest follow up was almost equal to those before start of ART.
Collapse
|