Radiation treatment in prostate cancer : balancing between tumor control and toxicity Heemsbergen, W.D.

Similar documents
UPDATE OF DUTCH MULTICENTER DOSE-ESCALATION TRIAL OF RADIOTHERAPY FOR LOCALIZED PROSTATE CANCER

The Phoenix Definition of Biochemical Failure Predicts for Overall Survival in Patients With Prostate Cancer

Department of Radiotherapy & Nuclear Medicine, National Cancer Institute, Cairo University, Cairo, Egypt.

Project approved by the Fondo de investigaciones Socio Sanitarias (FISS). Resolution dated June 8, Official State Gazette: June 17, 2004.

Outcomes Following Negative Prostate Biopsy for Patients with Persistent Disease after Radiotherapy for Prostate Cancer

Radiation dose has been reported to be an important determinant

The Dynamics of Dose Escalation of Radiotherapy for Localized Prostate Cancer. Abrahim Al-Mamgani

Vol. 36, pp , 2008 T1-3N0M0 : T1-3. prostate-specific antigen PSA. 68 Gy National Institutes of Health 10

SRO Tutorial: Prostate Cancer Clinics

HIGH DOSE RADIATION DELIVERED BY INTENSITY MODULATED CONFORMAL RADIOTHERAPY IMPROVES THE OUTCOME OF LOCALIZED PROSTATE CANCER

External Beam Radiation Therapy for Low/Intermediate Risk Prostate Cancer

Supported by M. D. Anderson Cancer Center physician investigator funds. We thank Gerald E. Hanks, MD, for help and guidance with this project.

New Technologies for the Radiotherapy of Prostate Cancer

Radiation Therapy for Prostate Cancer. Resident Dept of Urology General Surgery Grand Round November 24, 2008

Prostate Cancer. 3DCRT vs IMRT : Hasan Murshed

Prediction of toxicity in concurrent chemoradiation for non-small cell lung cancer Uijterlinde, W.I.

Prostate Cancer in comparison to Radiotherapy alone:

Jure Murgic 1, Matthew H Stenmark 1, Schuyler Halverson 1, Kevin Blas 1, Felix Y Feng 1,2 and Daniel A Hamstra 1,3*

The use of SpaceOAR hydrogel in dose-escalated prostate cancer radiotherapy and its impact on rectal dosimetry

Hormone Therapy for Prostate Cancer: Guidelines versus Clinical Practice

EORTC radiation Oncology Group Intergroup collaboration with RTOG EORTC 1331-ROG; RTOG 0924

Impact of the duration of hormonal therapy following radiotherapy for localized prostate cancer

Advances in Abdominal Aortic Aneurysm Care - Towards personalized, centralized and endovascular care van Beek, S.C.

Andrew K. Lee, MD, MPH Associate Professor Department tof fradiation Oncology M.D. Anderson Cancer Center

Hypofractionation for Prostate Cancer: the Present Luca Incrocci, MD PhD

Heterogeneity in high-risk prostate cancer treated with high-dose radiation therapy and androgen deprivation therapy

doi: /s (03) CLINICAL INVESTIGATION

Citation for published version (APA): Bartels, S. A. L. (2013). Laparoscopic colorectal surgery: beyond the short-term effects

When radical prostatectomy is not enough: The evolving role of postoperative

AMORE (Ablative surgery, MOulage technique brachytherapy and REconstruction) for childhood head and neck rhabdomyosarcoma Buwalda, J.

Radical Prostatectomy versus Intensity Modulated Radiation Therapy in the Management of Localized Prostate Cancer

Strategies of Radiotherapy for Intermediate- to High-Risk Prostate Cancer

High Risk Localized Prostate Cancer Treatment Should Start with RT

Overview of Radiotherapy for Clinically Localized Prostate Cancer

Proton beam therapy for prostate cancer. 1. What is the clinical effectiveness of proton beam therapy in the treatment of prostate cancer?

Michelle S Ludwig 1*, Deborah A Kuban 2, Xianglin L Du 4, David S Lopez 4, Jose-Miguel Yamal 5 and Sara S Strom 3

Monotherapy with Carbon Ion Radiation for Localized Prostate Cancer

Intensity Modulated Radiotherapy (IMRT) of the Prostate

Clinical Commissioning Policy Proposition: Proton Beam Therapy for Cancer of the Prostate

When PSA fails. Urology Grand Rounds Alexandra Perks. Rising PSA after Radical Prostatectomy

Salvage prostatectomy for post-radiation adenocarcinoma with treatment effect: Pathological and oncological outcomes

Comparison of external radiation therapy vs radical prostatectomy in lymph node positive prostate cancer patients

UvA-DARE (Digital Academic Repository) Towards image-guided radiotherapy of prostate cancer Smitsmans, M.H.P. Link to publication

Linac Based SBRT for Low-intermediate Risk Prostate Cancer in 5 Fractions: Preliminary Report of a Phase II Study with FFF Delivery

PSA bouncing after brachytherapy HDR and external beam radiation therapy: a study of 121 patients with minimum 5-years follow-up

Best Papers. F. Fusco

Locally advanced disease & challenges in management

Three-year outcomes of 324 prostate carcinoma patients treated with combination high-dose-rate brachytherapy and intensity modulated radiation therapy

Biochemical progression-free survival in localized prostate cancer patients treated with definitive external beam radiotherapy

Subject Index. Androgen antiandrogen therapy, see Hormone ablation therapy, prostate cancer synthesis and metabolism 49

Dose escalation with external beam therapy for

Clinical Study Clinical Results after High-Dose Intensity-Modulated Radiotherapy for High-Risk Prostate Cancer

Does RT favor RP in long term Quality of Life? Juanita Crook MD FRCPC Professor of Radiation Oncology University of British Columbia

Understanding the risk of recurrence after primary treatment for prostate cancer. Aditya Bagrodia, MD

Introduction ORIGINAL RESEARCH

Radiation Dose Escalation for Localized Prostate Cancer

The use of hormonal therapy with radiotherapy for prostate cancer: analysis of prospective randomised trials

New research in prostate brachytherapy

GRANDANGOLO: CA PROSTATA

NIH Public Access Author Manuscript World J Urol. Author manuscript; available in PMC 2012 February 1.

GUIDELINES ON PROSTATE CANCER

Michael J. Zelefsky MD a,, W. Robert Lee MD b, Anthony Zietman MD c, Najma Khalid MS d, Cheryl Crozier RN d, Jean Owen PhD d, J.

Citation for published version (APA): Wijkerslooth de Weerdesteyn, T. R. (2013). Population screening for colorectal cancer by colonoscopy

CLINICAL TRIALS Open clinical uro-oncology trials in Canada George Rodrigues, MD, Eric Winquist, MD

Citation for published version (APA): Donker, M. (2014). Improvements in locoregional treatment of breast cancer

J Clin Oncol 26: by American Society of Clinical Oncology INTRODUCTION

UvA-DARE (Digital Academic Repository) Improving aspects of palliative care for children Jagt, C.T. Link to publication

Development of a treatment planning protocol for prostate treatments using intensity modulated radiotherapy

Radiotherapy (RT) Protocol for Prostate Cancer

Outcomes of Radical Prostatectomy in Thai Men with Prostate Cancer

Debate: Whole pelvic RT for high risk prostate cancer??

Section: Therapy Effective Date: October 15, 2016 Subsection: Therapy Original Policy Date: December 7, 2011 Subject:

The Use of Conformal Radiotherapy and the Selection of Radiation Dose in T1 or T2 Prostate Cancer

UvA-DARE (Digital Academic Repository)

Time to failure after definitive therapy for prostate cancer: implications for importance of aggressive local treatment

VALUE AND ROLE OF PSA AS A TUMOUR MARKER OF RESPONSE/RELAPSE

Open clinical uro-oncology trials in Canada

2015 myresearch Science Internship Program: Applied Medicine. Civic Education Office of Government and Community Relations

OPTIMIZATION OF COLLIMATOR PARAMETERS TO REDUCE RECTAL DOSE IN INTENSITY-MODULATED PROSTATE TREATMENT PLANNING

BIOCHEMICAL RECURRENCE POST RADICAL PROSTATECTOMY

Locoregional treatment Session Oral Abstract Presentation Saulo Brito Silva

Acute toxicity profile in prostate cancer with conventional and hypofractionated treatment

Prostate Case Scenario 1

Open clinical uro-oncology trials in Canada Eric Winquist, MD, George Rodrigues, MD

Phase II study of FFF-SBRT in 5 fractions for low and intermediate risk prostate cancer

2/14/09. Why Discuss this topic? Managing Local Recurrences after Radiation Failure. PROSTATE CANCER Second Treatment

High-Dose Rate Temporary Prostate Brachytherapy. Original Policy Date

Prostate Cancer: 2010 Guidelines Update

failure (FBF) rates were calculated using the Phoenix definition.

Clinimetrics, clinical profile and prognosis in early Parkinson s disease Post, B.

Corporate Medical Policy

Future Directions in Prostate Cancer: The Case for Protons. John J. Coen, MD Helen & Harry Gray Cancer Center

A schematic of the rectal probe in contact with the prostate is show in this diagram.

Post Radical Prostatectomy Radiation in Intermediate and High Risk Group Prostate Cancer Patients - A Historical Series

Would SBRT Hypofractionated Approach Be as Good? Then Why Bother With Brachytherapy?

in 32%, T2c in 16% and T3 in 2% of patients.

Systematic Review of Brachytherapy & Proton Beam Therapy for Low-Risk Prostate Cancer: Preliminary Findings

Radiation with oral hormonal manipulation for non-metastatic, intermediate or high risk prostate cancer in men 70 and older or with comorbidities

Fecal Microbiota Transplantation: Clinical and experimental studies van Nood, E.

Evaluation of prognostic factors after radical prostatectomy in pt3b prostate cancer patients in Japanese population

Transcription:

UvA-DARE (Digital Academic Repository) Radiation treatment in prostate cancer : balancing between tumor control and toxicity Heemsbergen, W.D. Link to publication Citation for published version (APA): Heemsbergen, W. D. (2008). Radiation treatment in prostate cancer : balancing between tumor control and toxicity Amsterdam: The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital General rights It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons). Disclaimer/Complaints regulations If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: http://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible. UvA-DARE is a service provided by the library of the University of Amsterdam (http://dare.uva.nl) Download date: 15 Jul 2018

Dose-response in radiotherapy for localized prostate cancer: Results of the Dutch multicenter randomized phase III trial comparing 68 Gy with 78 Gy Stéphanie TH Peeters, Wilma D Heemsbergen, Peter CM Koper, Wim LJ van Putten, Annerie Slot, Michel FH Dielwart, Johannes MG Bonfrer, Luca Incrocci and Joos V Lebesque Journal of Clinical Oncology 2006, Volume 24, Issue 13, page 1990-1996.

ABSTRACT Purpose: To determine whether a dose of 78 Gy improves outcome compared with a conventional dose of 68 Gy for prostate cancer patients treated with three-dimensional conformal radiotherapy. Patients and methods: Between June 1997 and February 2003, stage T1b- T4 prostate cancer patients were enrolled onto a multicenter randomized trial comparing 68 Gy with 78 Gy. Patients were stratified by institution, age, (neo)adjuvant hormonal therapy (HT), and treatment group. Four treatment groups (with specific radiation volumes) were defined based on the probability of seminal vesicle involvement. The primary endpoint was freedom from failure (FFF). Failure was defined as clinical failure or biochemical failure, according to the American Society of Therapeutic Radiation Oncology definition. Other endpoints were: freedom from clinical failure (FFCF), overall survival (OS) and toxicity. Results: Median follow-up time was 51 months. Of the 669 enrolled patients, 664 were included in the analysis. HT was prescribed for 143 patients. FFF was significantly better in the 78 Gy arm compared with the 68 Gy arm (5 year FFF rate: 64 % v 54 %, respectively) with an adjusted hazard ratio of 0.74 (p = 0.02). No significant differences in FFCF or in OS were seen between the randomization arms. There was no difference in late genitourinary toxicity of Radiation Therapy Oncology Group and European Organization for Research and Treatment of Cancer (RTOG/EORTC) grade 2 or more, and a slightly higher non-significant incidence of late gastrointestinal RTOG/EORTC toxicity Grade 2 or more. Conclusions: This multicenter randomized trial shows a significantly improved FFF in prostate cancer patients treated with a higher dose of radiotherapy. Introduction Radiotherapy is one of the treatment options for localized prostate cancer, but with standard radiation doses (64 to 70 Gy), it is not always as effective as previously believed. One of the strategies to improve the efficiency of radiotherapy is increasing the dose. Such a dose-escalation has become possible with three-dimensional conformal radiotherapy (3D-CRT). This technology allows one to better conform the radiation fields to the target volume and to reduce the dose to the normal tissues. [1,2] Several Phase II 106

Improved outcome with High-dose RT trials have shown that, with 3D-CRT, higher than conventional doses are feasible. [3-7] A number of Phase III trials have been initiated, and three have published outcome results. [8-10] The first trial did not show an improved local control with higher doses. [8] Two recent trials found an improved freedom from failure (FFF) at the expense of an increase in overall gastrointestinal (GI) toxicity. [9,10] The use of (neo)adjuvant hormonal therapy (HT) was not allowed in any of these trials, whereas its use significantly improves overall survival (OS). [11] We performed a multicenter trial to test the hypothesis that a higher radiation dose of 78 Gy would lead to a higher FFF compared with 68 Gy in patients treated for localized prostate cancer with 3D-CRT, and we allowed the use of HT. We have already reported results on toxicity. [12,13] We found no significant differences between the randomization arms for acute and late overall genitourinary (GU) and GI toxicity. For some specific late toxicity items (rectal bleeding, fecal incontinence and nocturia) the incidences were significantly higher in the high-dose arm. In this article we present the first results on outcome of the trial, together with an update of overall GI and GU morbidity. Patients and methods Participants Four Dutch institutions enrolled patients onto the CKVO 96-10 randomized phase III trial. Pretreatment assessment included clinical examination (including digital rectal examination), transrectal ultrasound of the prostate, laboratory studies (CBC, creatinine, alkaline-phosphatase, gammaglutamyltransferases and initial prostate-specific-antigen (ipsa)), prostate biopsy with histological evaluation, bone scan, and a lymph node evaluation by a pelvic computed tomography scan or ultrasound, and/or surgical or cytological sampling when the estimated risk of involvement of the seminal vesicles (SV) was higher than 10 %. [14] The Abbott IMx immunoassay (Abbott, Abbott Park, IL) was recommended to measure ipsa levels. Eligibility criteria were adenocarcinoma of the prostate; all T-stages with ipsa < 60 μg/l, except T1a and well-differentiated (or Gleason Score < 5) T1b-c tumors with ipsa 4 μg/l; Karnofsky performance score of 80. Patients with metastases, with cytologically or histologically proven positive regional lymph nodes, on anticoagulants, with previous pelvic irradiation, and with previous malignant disease (other than basal cell carcinoma) were excluded. TNM-staging was scored according to the American Joint Committee on Cancer (AJCC) 1997 guidelines. 107

Patients were randomly assigned to either the conventional dose of 68 Gy, or the experimental dose of 78 Gy. Random assignment was performed with a minimization technique with stratification for treatment group (I, II, III and IV; see next section), institution (A, B, C and D), age ( 70 v > 70 years) and (neo)adjuvant hormonal therapy (HT) (yes v no). The protocol specified that HT was allowed, although not recommended until ongoing studies had defined groups that benefit from HT. Its prescription was left at the discretion of the treating physician. The Ethical Committee of each institution approved the protocol. All patients gave informed consent. Treatment The planning target volume (PTV) for radiation included the prostate with or without the SV (clinical target volumes, CTVs) with a margin of 10 mm during the first 68 Gy, and 5 mm (except towards the rectum, 0 mm) for the last 10 Gy in the high-dose arm. This 10 Gy could be regarded as a partial boost. Four radiation treatment groups were defined depending on the risk of tumor involvement of the SV. This risk was estimated according to Partin et al. based on T-Stage, ipsa and Gleason Score or differentiation grade. [14] (Table 1). T1b-T2a tumors with a risk of less than 10 %, 10 % to 25 %, and more than 25 % were included in groups I, II and III, respectively. T2b-T3a and T3b-T4 tumors were included in groups III and IV, respectively. For group I, the CTV was defined as the prostate only, and for group IV, it was defined as the prostate with SV. In groups II and III, the CTV included prostate and SV, and the SV were excluded from the CTV after 50 Gy and 68 Gy, respectively. Table 1. Treatment groups (I, II, III and IV). Gleason score 108 Differentiation ipsa (μg/l) T1b, T1c, T2a* T2b* T3a 0-4 4-10 10-20 20-60 0-60 2-4 Good I I I II III IV 5-7 Moderate I II II III III IV 8-10 Poor II III III III III IV Abbreviation: ipsa, initial prostate-specific antigen. *According to American Joint Committee on Cancer 1997 guidelines. All patients underwent computed tomography scan in treatment position (supine), and were treated with 3D-CRT, in daily fractions of 2 Gy. Each institution was allowed to use its own treatment technique. The dose was specified to the isocenter (International Commission on Radiation Units and Measurements reference point). [15] The dose to the PTVs was within 5 % T3b T4

Improved outcome with High-dose RT and +7 % of the prescribed dose and 99 % of the PTVs were treated to at least 95 % of the prescribed dose. The percentage of the rectum (or rectal wall) receiving 74 Gy should not have exceeded 40 % and the small bowel dose should not have been higher than 68 Gy. Protocol compliance has been checked in a quality control study. [16] All treatment plans were reviewed at the coordinating center using a dedicated database. [17] Follow-up visits were scheduled once every three months in the first year of follow-up, every four months in the second year, biannually in the following three years and yearly thereafter. Outcomes and Toxicity The primary endpoint was FFF. We defined failure as biochemical or clinical failure whichever was first. A clinical failure included local relapse (defined as palpable and/or biopsy-proven progression), regional relapse, metastases, or initiation of salvage HT. Prostate biopsies were not systematically performed. Biochemical failure was defined according to the definition of the ASTRO and was considered three consecutive increases in PSA level after a nadir. [18] The date was defined as midway between the last non-increasing value and the first rise of PSA. A second analysis without backdating was performed, because of concerns that backdating may influence the failure rate. [19,20] Time to failure was calculated from the date of random assignment with patients censored at the date of the last PSA measurement or death. Other endpoints were freedom from clinical failure (FFCF), OS, and toxicity. In this article, we present an update of GI and GU toxicity scored according to slightly modified RTOG/EORTC scoring criteria. [12] Besides the radiation treatment groups, patients were retrospectively divided into three prognostic risk groups as published in the literature (low, intermediate and high risk). They were defined according to the single-factor definition of Chism et al. [21] The low-risk group included patients with Stages T1-T2 and Gleason Score 6 and ipsa 10 μg/l. The high-risk group contained Stages T3-T4 or Gleason Score 8 or ipsa > 20 μg/l. The intermediate-risk group included patients that did not fulfill the criteria of the low- or high-risk group. For 93 patients, the differentiation grade only was available. Forty-two of these patients had stage T3-T4 or ipsa > 20 μg/l, and were therefore high-risk patients. For the remaining 51 patients, the differentiation grade was used to assign a risk group; well-differentiated, moderately differentiated and poorly differentiated tumors were considered to be Gleason Scores 2 to 6, 7 and 8 to 10, respectively. 109

Statistical analysis The study was designed to have an 80 % power of detecting a difference in FFF of 10 % between patients treated to 68 and 78 Gy at the 5 % level of significance (two sided). Analyses were performed by intention to treat. Cox proportional hazards regression was used to analyze differences (expressed in hazard ratios) in FFF, FFCF and OS between both arms. These analyses were adjusted for the stratification factors (treatment group, institution, age and HT). Survival curves were estimated by the Kaplan Meier technique. To analyze the dose effect in the three risk groups and in the three HT groups (no, short-term and long-term HT) the hazard ratios were estimated by stratified Log Rank analysis. [22] Results Between June 1997 and February 2003, 669 patients were recruited. Five patients were excluded from the analysis (Fig. 1); three did not fulfill the eligibility criteria, and two went off study before start of radiotherapy. Therefore we analyzed 664 patients: 331 were allocated to 68 Gy and 333 to 78 Gy. Follow-up data were reported until May 2005. The median follow-up was 50.7 months (range 9.6 to 94.2 months). Patient and tumor baseline characteristics were well balanced between both arms (Table 2). The lowdose arm included slightly more patients with high-risk features, but the four treatment groups, based on risk of SV invasion, and the three risk groups were well-balanced between both arms. (Neo)adjuvant hormonal therapy (HT) was prescribed in two hospitals for 143 patients, and prescribed predominantly to high-risk patients (n = 125), and rarely to intermediate-risk (n = 13) or low-risk patients (n = 5). HT was initiated 0 to 7 months before radiotherapy for 6 months (short-term HT, institution B) or 3 years (long-term HT, institution A). Generally, a luteinizing hormone-releasing hormone agonist, preceded by a short course of an anti-androgen was prescribed. The use of short-term and long-term HT was well balanced (Table 2). Several treatment techniques were used: fourfield technique (n = 70, institution C), three-field technique using one anterior and two lateral wedged fields (n = 553), and intensity-modulated radiotherapy (simultaneous integrated boost [23] for 41 patients in the highdose arm, institution B). In the low-dose arm all patients received the prescribed 68 Gy, whereas in the high-dose arm, 36 patients received a dose between 68 and 76 Gy, and one patient died during treatment from a disease-unrelated cause. In 19 patients the dose was lowered to meet the rectal (n = 7) or small bowel (n = 12) dose constraints. Other reasons were 110

Improved outcome with High-dose RT technical problems (n = 3), patient s request (n = 5), and acute toxicity (n = 9). Only 3 of these 14 latter patients had severe acute GU toxicity. Fig. 1. Trial profile. 669 patients included 332 patients allocated to 68 Gy 337 patients allocated to 78 Gy 1 not eligible 2 not eligible 2 not treated according to trial 331 patients in main analysis 333 patients in main analysis Outcome At the time of assessment, failure had occurred in 136 patients in the lowdose, and in 107 patients in the high-dose arm. There were more biochemical failures in the 68 Gy arm (n = 118) compared with the 78 Gy arm (n = 85) (Table 3). For three patients, the cause of death could not be traced, and therefore, we added these patients to the clinical failure group. FFF was significantly higher in the high-dose arm compared with the low-dose arm (Log Rank p = 0.01), with an adjusted hazard ratio of 0.74 (95 % CI 0.58-0.96, p = 0.02). The 5-year FFF Kaplan Meier estimates were 64 % in the high-dose, and 54 % in the low-dose arm (Fig. 2A); these figures were 66 % and 53 % when biochemical failure was defined using the ASTRO definition without backdating (Log Rank p = 0.02; Fig. 2B). Clinical failure was seen in 66 and 69 patients in the low-dose and high-dose arm, respectively. Approximately half of the patients had a locoregional or distant relapse (Table 4). Salvage HT was preceded by a biochemical failure for most of the patients. There was no significant difference in FFCF between both arms, with 5-year FFCF estimates of 76 % in both arms. Forty-nine patients in the low-dose and 43 patients in the highdose arm had died. The number of patients who died from prostate cancer was similar in both arms (20 and 21 patients in the low- and high-dose arm, respectively). No significant differences were seen between both arms in terms of OS. The 5-year OS incidences were 82 % and 83 % in the low- and high-dose arm, respectively. 111

Table 2. Baseline characteristics. Variable 68 Gy (n=331) 78 Gy (n=333) Mean Age (range) (years) 68.6 (50-83) 68.8 (49-84) Institution A B C D 201 86 35 9 61 % 26 % 11 % 3 % 203 85 35 10 61 % 26 % 11 % 3 % Hormonal therapy Treatment Group Differentiation group (Gleason score) (cfr table 1) Tumor stage Short-term Long-term I II III IV Good (2-4) Moderate (5-7) Poor (8-10) T1 T2 T3 T4 ipsa (μg/l) 4 4-10 10-20 20-60 Risk groups Low Intermediate High 73 35 38 55 65 156 55 105 170 56 57 151 116 7 25 95 125 86 56 90 185 Abbreviations: ipsa, initial prostate-specific antigen. 22 % 11 % 11 % 17 % 20 % 47 % 17 % 32 % 51 % 17 % 17 % 45 % 35 % 2 % 8 % 29 % 38 % 26 % 17 % 27 % 56 % 70 29 41 52 67 163 51 94 194 45 67 141 123 2 19 119 125 70 64 92 177 21% 9 % 12 % 16 % 20 % 49 % 15 % 28 % 59 % 14 % 21 % 42 % 37 % 1 % 6 % 36 % 38 % 21 % 19 % 28 % 53 % Table 3. First failure type. Type of progression Biochemical failure Clinical failure Locoregional or DM Salvage HT Death* 118 18 9 7 2 68 Gy (n=331) 36 % 5 % 3 % 2 % 0.6 % 85 22 13 8 1 78 Gy (n=333) 26 % 7 % 4 % 2 % 0.3 % Total (n=664) 203 40 22 15 3 Abbreviations: DM, distant metastasis; HT, hormonal treatment. * Patients for whom death from prostate cancer could not be excluded. 31 % 6 % 3 % 2 % 0.5 % 112

Improved outcome with High-dose RT Table 4: Clinical failures for the endpoint freedom from clinical failure. Type of Clinical failure 68 Gy (n=66) 78 Gy (n=69) Locoregional or distant metastases - Local - Regional - Distant Salvage HT - Preceded by biochemical failure - Median time to start Abbreviations: HT, hormonal treatment. 32 13 3 20 34 27 30.7 months 38 7 8 28 31 23 32.2 months Subgroup analyses A lower failure rate was seen in the high-dose arm in the intermediate- and high-risk group, with a difference in relative number of failures of 15 % and 8 %, respectively (Fig. 3). The 95 % CI demonstrated that this difference was significant (hazard ratio = 0.6) only for the intermediate-risk group. There was no apparent benefit for the low-risk patients. However, a test for interaction between random assignment arm and prognostic risk group for probability of FFF was not significant (p = 0.3). The test for interaction between random assignment arm and the three HT-groups (no, short-term and long-term HT) was not significant either (p = 0.4). Toxicity Late GI toxicity grade 2 was slightly higher in the high-dose arm, but this difference was not significant (5-year toxicity rate: 32 % vs. 27 %; Log Rank p = 0.2). No differences were seen between both arms for late GI Grade 3 (5 % vs. 4 %; p = 0.4), late GU toxicity Grade 2 (39 % vs. 41 %; p = 0.6) and GU Grade 3 (13 % vs. 12 %; p = 0.6). 113

A 1.0 0.8 EXP 78 Cumulative Incidence 0.6 0.4 CONV 68 0.2 0.0 0 1 2 3 4 5 Time from randomisation (years) N at risk CONV 68 331 290 208 134 80 49 EXP 78 333 293 236 163 109 65 B 1.0 N F CONV 68 331 136 EXP 78 333 107 Log-rank p = 0.01 Abbreviations: EXP = experimental; CONV = conventional; F = failures. 0.8 EXP 78 Cumulative Incidence 0.6 0.4 CONV 68 0.2 N F CONV 68 331 136 EXP 78 333 107 Log-rank p = 0.02 0.0 0 1 2 3 4 5 Time from randomisation (years) N at risk CONV 68 331 319 272 180 106 64 EXP 78 333 316 264 195 130 76 Fig. 2. Kaplan-Meier estimates of freedom from failure by treat-ment arm, where biochemical failure was defined according to the ASTRO definition (A) with and (B) without backdating. 114

Improved outcome with High-dose RT Failure / Patients Risk 78 Gy 68 Gy HR & 95% CI Low Intermediate High Total 10 / 64 19 / 92 78 / 177 107 / 333 (32%) 7 / 56 32 / 90 97 / 185 136 / 331 (41%) Fig. 3: Number of failures (biochemical/clinical), and hazard ratios (HRs; with 95% CIs) showing the effect of the randomization arm on FFF for the low, intermediate and high risk groups, as well as for the whole patient group. Discussion This study shows that, for localized prostate cancer, the use of higher radiation doses significantly improved the FFF probability. The risk of failure was reduced from 46 % to 36 % at 5 years (Fig. 2A). Failure rates were lower in the high-dose arm both in the intermediate- and high-risk groups, but not in the low-risk patients (Fig. 3). However, this trial was not powered to perform these subgroup analyses, and the low-risk group included only a small number of patients. Furthermore, the test for interaction showed that there was no significant difference in dose effect between the three risk groups. Therefore we can not exclude that low-risk patients also benefit from dose escalation. Because we have shown that a high dose was a significant prognostic factor for FFF, and a number of patients in the high-dose arm received a lower than prescribed dose, we also analyzed all patients by the actually given dose (low-dose group: mean dose 67.9 Gy; range: 16 to 72 Gy v high-dose group: mean dose 77.9 Gy; range: 74 to 78 Gy), instead of by intention to treat as in the main analysis. Comparison of these two groups resulted in an even larger difference in FFF (p = 0.001; results not shown). Two other Phase III trials have found a dose response for freedom from failure. [9,10] The M.D. Anderson Cancer Centre trial compared 70 Gy with 115

78 Gy in 305 stage T1-T3 prostate cancer patients and found a significantly improved FFF. A preliminary report showed an absolute benefit of 10 % at 5 years, which is similar to our results. [24] In a second analysis with an additional follow-up of 20 months the benefit at 6 years was 6 %. [9] An older phase III trial, carried out by the Massachusetts General Hospital, compared 67.2 Gy with 75.6 Cobalt Gray Equivalent (GyE) in 202 high-risk prostate cancer patients with stages T3-4N0-2. [8] They used a proton boost to increase the dose. No significant difference was found in local control in the entire cohort. Subsequently, they performed another phase III trial together with the Loma Linda Medical Center. That study included 393 patients and compared 70.2 GyE with 79.2 GyE, again using a proton boost and including only patients with stages T1-T2b disease. [10] A large advantage in freedom from biochemical failure was noted for patients treated to high doses, and this difference held true for low-risk as well as for higher risk patients. Most of the non-randomized studies found a dose-response for intermediate-risk patients [25-27], some also for high-risk patients [25,26], but only few studies detected a benefit for low-risk patients. [28,29] Of note is that the curves for the two dose levels in our study started to separate after approximately 1.5 years when biochemical failure was defined using the ASTRO definition (Fig 2A), and after 3 years without backdating (Fig 2B). The patients with an early failure were mainly high-risk patients who developed a failure outside the prostate (results not shown). A comparable lag period before separation of the two curves was seen in the study of Zietman et al. [10] We found no difference in FFCF or OS between the two dose levels. We need a longer follow-up to show a dose response in FFCF given that biochemical failure has been shown to be an early surrogate for clinical failure. [30-32] Because Kestin et al. found that the median time between the third consecutive PSA rise and clinical failure was 1.4 years, it is not surprising that we did not find a difference in FFCF yet, given that our curves separated after 3 years (Fig 2B). A major difference with the three other Phase III trials is that we allowed (neo)adjuvant HT. [8-10] After cessation of HT, a transient increase in PSA may occur due to recovery of prostate tissue from testosterone suppression. This may lead to false-positive results with the ASTRO definition. [33] Therefore, discussion has arisen whether another definition of PSA failure should be used for patients treated with HT. The results of studies that compared several definitions of biochemical failure in patients treated with HT vary with conflicting results. [33-35] Therefore, we repeated the main analysis with a proposed alternative definition of failure (nadir plus 2 μg/l), and found that FFF was higher in the high-dose arm versus the lowdose arm (at 5 years 67 % vs. 61 %), but this difference was not significant 116

Improved outcome with High-dose RT (Log Rank p = 0.2). [20,35] Another criticism on the ASTRO definition is that it was designed based on data where PSA levels below 0.5 μg/l could not be measured. [36] For that reason we also analyzed FFF with data where PSA-values smaller than 0.5 μg/l were set to 0.5 μg/l. We found that the differences in FFF between both arms were still significant (results not shown). An advantage of this latter definition is that some of the small PSA bounces are left out. However, whatever the shortcomings of the ASTRO definition are, they probably occur in both arms because of the random assignment. Nevertheless, increasing the dose is not without consequences with regard to toxicity. In this paper we presented an update of overall late gastrointestinal and genitourinary toxicity and found no significant differences between both randomized arms, which is in accordance with our earlier results. [12] However, for the more specific symptoms late rectal bleeding and fecal incontinence, a higher radiation dose resulted in significantly higher incidences. [12,13] In contrast to our first report, nocturia was no longer significantly higher in the high-dose arm (data not shown). [12] Although a higher treatment dose resulted in increased GI toxicity, the incidences were still acceptable. We also expect that, with the use of intensity-modulated radiotherapy for all patients, the complication rates will decrease. [37] Conclusions In summary, we have shown that prostate cancer patients benefited from an increase of the dose by 10 Gy in terms of freedom from failure, at the cost of slightly higher, but acceptable, incidences of late rectal bleeding and fecal incontinence. This result supports the use of a higher radiation dose in patients with localized prostate cancer. 117

References 1. Koper PC, Jansen P, van Putten W, et al. Gastro-intestinal and genito-urinary morbidity after 3D conformal radiotherapy of prostate cancer: observations of a randomized trial. Radiother Oncol 2004;73:1-9. 2. Dearnaley DP, Khoo VS, Norman AR, et al. Comparison of radiation side-effects of conformal and conventional radiotherapy in prostate cancer: a randomised trial. Lancet 1999;353:267-272. 3. Michalski JM, Purdy JA, Winter K, et al. Preliminary report of toxicity following 3D radiation therapy for prostate cancer on 3DOG/RTOG 9406. Int J Radiat Oncol Biol Phys 2000;46:391-402. 4. Leibel SA, Heimann R, Kutcher GJ, et al. Three-dimensional conformal radiation therapy in locally advanced carcinoma of the prostate: preliminary results of a phase I doseescalation study. Int J Radiat Oncol Biol Phys 1994;28:55-65. 5. Boersma LJ, van den Brink M, Bruce AM, et al. Estimation of the incidence of late bladder and rectum complications after high-dose (70-78 Gy) conformal radiotherapy for prostate cancer, using dose-volume histograms. Int J Radiat Oncol Biol Phys 1998;41:83-92. 6. Hanks GE, Schultheiss TE, Hanlon AL, et al. Optimization of conformal radiation treatment of prostate cancer: report of a dose escalation study. Int J Radiat Oncol Biol Phys 1997;37:543-550. 7. Bey P, Carrie C, Beckendorf V, et al. Dose escalation with 3D-CRT in prostate cancer: French study of dose escalation with conformal 3D radiotherapy in prostate cancerpreliminary results. Int J Radiat Oncol Biol Phys 2000;48:513-517. 8. Shipley WU, Verhey LJ, Munzenrider JE, et al. Advanced prostate cancer: the results of a randomized comparative trial of high dose irradiation boosting with conformal protons compared with conventional dose irradiation using photons alone. Int J Radiat Oncol Biol Phys 1995;32:3-12. 9. Pollack A, Zagars GK, Starkschall G, et al. Prostate cancer radiation dose response: results of the M. D. Anderson phase III randomized trial. Int J Radiat Oncol Biol Phys 2002;53:1097-1105. 10. Zietman AL, DeSilvio ML, Slater JD, et al. Comparison of conventional-dose vs. high-dose conformal radiation therapy in clinically localized adenocarcinoma of the prostate: a randomized controlled trial. JAMA 2005;294:1233-1239. 11. Bolla M, Collette L, Blank L, et al. Long-term results with immediate androgen suppression and external irradiation in patients with locally advanced prostate cancer (an EORTC study): a phase III randomised trial. Lancet 2002;360:103-106. 12. Peeters ST, Heemsbergen WD, van Putten WL, et al. Acute and late complications after radiotherapy for prostate cancer: results of a multicenter randomized trial comparing 68 Gy to 78 Gy. Int J Radiat Oncol Biol Phys 2005;61:1019-1034. 13. Peeters ST, Lebesque JV, Heemsbergen WD, et al. Localized volume effects for late rectal and anal toxicity after radiotherapy for prostate cancer. Int J Radiat Oncol Biol Phys 2006;64:1151-61. 14. Partin AW, Yoo J, Carter HB, et al. The use of prostate specific antigen, clinical stage and Gleason score to predict pathological stage in men with localized prostate cancer. J Urol 1993;150:110-114. 15. International Commission on Radiation Units and Measurements (ICRU) Report No. 50: Prescribing, recording and reporting photon beam therapy. Bethesda, MD: ICRU. 1993 16. Rasch C, Remeijer P, Koper PC, et al. Comparison of prostate cancer treatment in two institutions: a quality control study. Int J Radiat Oncol Biol Phys 1999;45:1055-1062. 17. Hoogeman MS, van Herk M, and Zijp L. The design and implementation of a multicenter volumetric and dosimetric database. Berlin Springer 82-84, 2000. 18. ASTRO: Consensus statement: guidelines for PSA following radiation therapy. American Society for Therapeutic Radiology and Oncology Consensus Panel. Int J Radiat Oncol Biol Phys 1997;37:1035-41. 19. Connell PP, Ignacio L, McBride RB, et al. Caution in interpreting biochemical control rates after treatment of prostate cancer: length of follow-up influences results. Urology 1999;54:875-79. 118

Improved outcome with High-dose RT 20. Thames H, Kuban D, Levy L, et al. Comparison of alternative biochemical failure definitions based on clinical outcome in 4839 prostate cancer patients treated by external beam radiotherapy between 1986 and 1995. Int J Radiat Oncol Biol Phys 2003;57:929-43. 21. Chism DB, Hanlon AL, Horwitz EM, et al. A comparison of the single and double factor high-risk models for risk assignment of prostate cancer treated with 3D conformal radiotherapy. Int J Radiat Oncol Biol Phys 2004;59:380-85. 22. Early Breast Cancer Trialists' Collaborative Group. Effects of adjuvant tamoxifen and of cytotoxic therapy on mortality in early breast cancer. An overview of 61 randomized trials among 28,896 women. N Engl J Med 1988;319:1681-92. 23. Bos LJ, Damen EM, de Boer RW et al. Reduction of rectal dose by integration of the boost in the large-field treatment plan for prostate irradiation. Int J Radiat Oncol Biol Phys 2002;52:254-65. 24. Pollack A, Zagars GK, Smith LG, et al. Preliminary results of a randomized radiotherapy dose-escalation study comparing 70 Gy with 78 Gy for prostate cancer. J Clin Oncol 2000;18:3904-11. 25. Zelefsky MJ, Leibel SA, Gaudin PB, et al. Dose escalation with three-dimensional conformal radiation therapy affects the outcome in prostate cancer. Int J Radiat Oncol Biol Phys 1998;41:491-500. 26. Hanks GE, Hanlon AL, Pinover WH, et al. Dose selection for prostate cancer patients based on dose comparison and dose response studies. Int J Radiat Oncol Biol Phys 2000;46:823-32. 27. Symon Z, Griffith KA, McLaughlin PW, et al. Dose escalation for localized prostate cancer: substantial benefit observed with 3D conformal therapy. Int J Radiat Oncol Biol Phys 2003;57:384-90. 28. Lyons JA, Kupelian PA, Mohan DS, et al. Importance of high radiation doses (72 Gy or greater) in the treatment of stage T1-T3 adenocarcinoma of the prostate. Urology 2000;55:85-90. 29. Kupelian PA, Buchsbaum JC, Reddy CA, et al. Radiation dose response in patients with favorable localized prostate cancer (Stage T1-T2, biopsy Gleason < or = 6, and pretreatment prostate-specific antigen < or = 10). Int J Radiat Oncol Biol Phys 2001;50:621-25. 30. Jacob R, Hanlon AL, Horwitz EM, et al. The relationship of increasing radiotherapy dose to reduced distant metastases and mortality in men with prostate cancer. Cancer 2004;100:538-43. 31. Vicini FA, Kestin LL, and Martinez AA. The correlation of serial prostate specific antigen measurements with clinical outcome after external beam radiation therapy of patients for prostate carcinoma. Cancer 2000;88:2305-18. 32. Kestin LL, Vicini FA, and Martinez AA. Practical application of biochemical failure definitions: what to do and when to do it. Int J Radiat Oncol Biol Phys 2002;53:304-15. 33. Zietman AL, Christodouleas JP, and Shipley WU. PSA bounces after neoadjuvant androgen deprivation and external beam radiation: impact on definitions of failure. Int J Radiat Oncol Biol Phys 2005;62:714-18. 34. Pickles T, Kim-Sing C, Morris WJ, et al. Evaluation of the Houston biochemical relapse definition in men treated with prolonged neoadjuvant and adjuvant androgen ablation and assessment of follow-up lead-time bias. Int J Radiat Oncol Biol Phys 2003;57:11-18. 35. Buyyounouski MK, Hanlon AL, Eisenberg DF, et al. Defining Biochemical Failure after Radiotherapy with and without Androgen Deprivation for Prostate Cancer. Int J Radiat Oncol Biol Phys 2005;63:1455-82. 36. Zietman AL and Shipley WU. In response to Pickles. Int J Radiat Oncol Biol Phys 1999;43:700. 37. Zelefsky MJ, Fuks Z, Happersett L, et al. Clinical experience with intensity modulated radiation therapy (IMRT) in prostate cancer. Radiother Oncol 2000;55:241-49. 119

120