1
|
Turkbey B, Oto A, Allen BC, Akin O, Alexander LF, Ari M, Froemming AT, Fulgham PF, Gettle LM, Maranchie JK, Rosenthal SA, Schieda N, Schuster DM, Venkatesan AM, Lockhart ME. ACR Appropriateness Criteria® Post-Treatment Follow-up of Prostate Cancer: 2022 Update. J Am Coll Radiol 2023; 20:S164-S186. [PMID: 37236741 DOI: 10.1016/j.jacr.2023.02.012] [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: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 05/28/2023]
Abstract
Prostate cancer has a wide spectrum ranging between low-grade localized disease and castrate-resistant metastatic disease. Although whole gland and systematic therapies result in cure in the majority of patients, recurrent and metastatic prostate cancer can still occur. Imaging approaches including anatomic, functional, and molecular modalities are continuously expanding. Currently, recurrent and metastatic prostate cancer is grouped in three major categories: 1) Clinical concern for residual or recurrent disease after radical prostatectomy, 2) Clinical concern for residual or recurrent disease after nonsurgical local and pelvic treatments, and 3) Metastatic prostate cancer treated by systemic therapy (androgen deprivation therapy, chemotherapy, immunotherapy). This document is a review of the current literature regarding imaging in these settings and the resulting recommendations for imaging. The American College of Radiology Appropriateness Criteria are evidence-based guidelines for specific clinical conditions that are reviewed annually by a multidisciplinary expert panel. The guideline development and revision include an extensive analysis of current medical literature from peer reviewed journals and the application of well-established methodologies (RAND/UCLA Appropriateness Method and Grading of Recommendations Assessment, Development, and Evaluation or GRADE) to rate the appropriateness of imaging and treatment procedures for specific clinical scenarios. In those instances where evidence is lacking or equivocal, expert opinion may supplement the available evidence to recommend imaging or treatment.
Collapse
Affiliation(s)
- Baris Turkbey
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Aytekin Oto
- Panel Chair, University of Chicago, Chicago, Illinois
| | - Brian C Allen
- Panel Vice-Chair, Duke University Medical Center, Durham, North Carolina
| | - Oguz Akin
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Mim Ari
- The University of Chicago, Chicago, Illinois, Primary care physician
| | | | - Pat F Fulgham
- Urology Clinics of North Texas, Dallas, Texas; American Urological Association
| | | | | | - Seth A Rosenthal
- Sutter Medical Group, Sacramento, California; Commission on Radiation Oncology
| | - Nicola Schieda
- Ottawa Hospital Research Institute and the Department of Radiology, The University of Ottawa, Ottawa, Ontario, Canada
| | - David M Schuster
- Emory University, Atlanta, Georgia; Commission on Nuclear Medicine and Molecular Imaging
| | | | - Mark E Lockhart
- Specialty Chair, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
2
|
Onal C, Erbay G, Oymak E, Cem Guler O. The impact of the apparent diffusion coefficient for the early prediction of the treatment response after definitive radiotherapy in prostate cancer patients. Radiother Oncol 2023; 184:109677. [PMID: 37084886 DOI: 10.1016/j.radonc.2023.109677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/06/2023] [Accepted: 04/12/2023] [Indexed: 04/23/2023]
Abstract
PURPOSE We assessed early changes in apparent diffusion coefficient (ADC) and serum prostate specific antigen (PSA) values after definitive radiotherapy (RT) without androgen deprivation treatment in low- and intermediate-risk prostate cancer (PC) patients. MATERIALS AND METHODS The clinical data and ADC parameters of 229 PC patients were retrospectively evaluated. Pre-treatment and post-treatment serum PSA and primary tumor ADC values were calculated. Post-treatment DW-MRI was performed median 4.1 months after completion of definitive RT. The prognostic factors predicting freedom from biochemical failure (FFBF) and progression-free survival (PFS) were analyzed using univariable and multivariable analyses. RESULTS With a median follow-up time of 80.8 months, the 5-year FFBF and PFS rates were 95.9% and 89.3%, respectively. Eleven patients (4.8%) had PSA relapse, with a median of 34.4 months after the completion of RT. A statistically significant difference in post-treatment ADC values was noted between patients with and without recurrence (0.94 ± 0.07 vs. 1.10 ± 0.20 × 10-3 mm2/sec; p < 0.001). Patients with a Gleason score (GS) of 6 and low-risk disease had significantly higher post-treatment tumor ADC and PSA levels than patients with a GS of 7 and intermediate-risk disease. The 5-year FFBF rate in patients with tumor ADC ≤ 0.96 × 10-3 mm2/sec was significantly lower than patients with tumor ADC > 0.96 × 10-3 mm2/sec (85.5% vs. 100; p < 0.001). In the multivariable analysis, a lower ADC value, GS 4 + 3 and intermediate-risk disease were independent predictors of worse FFBF. In the multivariate analysis, a lower post-treatment ADC value and a GS of 4+3 were significant prognostic factors for a lower PFS. CONCLUSION These findings suggest that the post-treatment tumor ADC value could be used for early treatment response evaluation after definitive RT in PC patients.
Collapse
Affiliation(s)
- Cem Onal
- Department of Radiation Oncology, Baskent University Faculty of Medicine, Dr. Turgut Noyan Research and Treatment Center, Adana, Turkey; Department of Radiation Oncology, Baskent University Faculty of Medicine, Ankara, Turkey.
| | - Gurcan Erbay
- Department of Radiology, Baskent University Faculty of Medicine, Dr. Turgut Noyan Research and Treatment Center, Adana, Turkey
| | - Ezgi Oymak
- Division of Radiation Oncology, Iskenderun Gelisim Hospital, Hatay, Turkey
| | - Ozan Cem Guler
- Department of Radiation Oncology, Baskent University Faculty of Medicine, Dr. Turgut Noyan Research and Treatment Center, Adana, Turkey
| |
Collapse
|
3
|
Prognostic value of PSA bounce after definitive radiotherapy revisited. Int J Clin Oncol 2021; 27:411-417. [PMID: 34677737 DOI: 10.1007/s10147-021-02053-0] [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: 07/22/2021] [Accepted: 10/07/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Prostate-specific antigen (PSA) bounce after definitive radiotherapy has been reported as a predictor of improved biochemical recurrence-free survival (BCRFS). We revisited this phenomenon to confirm its clinical impact on oncological outcomes in patients with long-term follow-up who were free of biochemical recurrence (BCR) at least 3 years after treatment. MATERIALS AND METHODS A total of 541 patients with localized, intermediate-risk prostate cancer underwent low-dose rate brachytherapy with iodine-125 seeds with or without supplemental external beam radiotherapy in combination. Neoadjuvant hormonal therapy was administered to 273 patients (50.5%) with a median duration of 3 months (range 1-108 months). PSA bounce was defined as ≥ 0.2 ng/ml increase above the interval PSA nadir, followed by a decrease below that value. RESULTS The median age was 69 years (range 49-90 years). The median follow-up duration was 102 months (range 36-205 months). One-hundred and fifty patients (27.7%) had PSA bounce with a median magnitude of 0.47 ng/ml (range 0.2-3.19 ng/ml). Age was significantly associated with the occurrence of PSA bounce [age: hazard ratio (HR) 0.95, 95% confidence interval (CI) 0.93-0.98]. It was found to be independently associated with a decreased risk for BCR (HR 0.29; 95% CI 0.12-0.69) and clinical progression (HR 0.44; 95% CI 0.95-0.98). CONCLUSION PSA bounce indicated a favorable BCRFS and clinical progression-free survival in patients who had been free of BCR for at least 3 years after definitive radiotherapy.
Collapse
|
4
|
Kishan AU. PSA bounce, prognosis, and clues to the radiation response. Prostate Cancer Prostatic Dis 2021; 24:937-938. [PMID: 34007012 DOI: 10.1038/s41391-021-00387-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 04/23/2021] [Accepted: 04/28/2021] [Indexed: 11/09/2022]
Affiliation(s)
- Amar U Kishan
- Departments of Radiation Oncology and Urology, University of California, Los Angeles, CA, USA.
| |
Collapse
|
5
|
Takakusagi Y, Oike T, Kano K, Anno W, Tsuchida K, Mizoguchi N, Serizawa I, Yoshida D, Katoh H, Kamada T. Prostate-specific antigen dynamics after neoadjuvant androgen-deprivation therapy and carbon ion radiotherapy for prostate cancer. PLoS One 2020; 15:e0241636. [PMID: 33156884 PMCID: PMC7647067 DOI: 10.1371/journal.pone.0241636] [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: 06/18/2020] [Accepted: 10/16/2020] [Indexed: 11/18/2022] Open
Abstract
Background This study aimed to explain the dynamics of prostate-specific antigen (PSA) levels in patients with prostate cancer who were treated with carbon ion radiotherapy (CIRT) and neoadjuvant androgen-deprivation therapy (ADT). Methods Eighty-five patients with intermediate-risk prostate cancer who received CIRT and neoadjuvant ADT from December 2015 to December 2017 were analyzed in the present study. The total dose of CIRT was set at 51.6 Gy (relative biological effectiveness) delivered in 12 fractions over 3 weeks. The PSA bounce was defined as a ≥0.4 ng/ml increase of PSA levels from the nadir, followed by any decrease. PSA failure was defined using the Phoenix criteria. Results The median patient age was 68 (range, 48–81) years. The median follow-up duration was 33 (range, 20–48) months. The clinical T stage was T1c, T2a, and T2b in 27, 44, and 14 patients, respectively. The Gleason score was 6 in 3 patients and 7 in 82 patients. The median pretreatment PSA level was 7.37 (range, 3.33–19.0) ng/ml. All patients received neoadjuvant ADT for a median of 6 (range, 2–117) months. PSA bounces were observed in 39 patients (45.9%), occurring a median of 12 (range, 6–30) months after CIRT. PSA failure was observed in eight patients (9.4%), occurring a median of 21 (range, 15–33) months after CIRT. The 3-year PSA failure-free survival rate was 88.5%. No clinical recurrence was observed during the follow-up period. Younger age and lower T stage were significant predictors of PSA bounce. Younger age was a significant predictor of PSA failure. Conclusions In this study, we identified the significant predictors of the occurrence of PSA bounce and failure. Further follow-up is needed to reveal the clinical significance of PSA dynamics.
Collapse
Affiliation(s)
- Yosuke Takakusagi
- Department of Radiation Oncology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Takahiro Oike
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Kio Kano
- Department of Radiation Oncology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Wataru Anno
- Department of Radiation Oncology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Keisuke Tsuchida
- Department of Radiation Oncology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Nobutaka Mizoguchi
- Department of Radiation Oncology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Itsuko Serizawa
- Department of Radiation Oncology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Daisaku Yoshida
- Department of Radiation Oncology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Hiroyuki Katoh
- Department of Radiation Oncology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Tadashi Kamada
- Department of Radiation Oncology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| |
Collapse
|
6
|
Time to achieve a prostate-specific antigen nadir of ≤0.2 ng/mL and related factors after permanent prostate brachytherapy. Brachytherapy 2020; 20:29-37. [PMID: 32814663 DOI: 10.1016/j.brachy.2020.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 11/24/2022]
Abstract
PURPOSE The purpose of this study was to identify the time to achieve a prostate-specific antigen (PSA) nadir of ≤0.2 ng/mL and the related factors to achieve this goal. MATERIALS AND METHODS We retrospectively reviewed 2218 Japanese prostate cancer patients who received 125I brachytherapy with or without external beam radiotherapy between 2003 and 2013 at one institution. Among them, patients followed up for ≥72 months and without luteinizing hormone-releasing hormone (LH-RH) agonist/antagonist were included (total of 1089 patients). The time to a PSA nadir of ≤0.2 ng/mL (months) was defined as the time between the date of implantation and the first time the lowest PSA value reached ≤0.2 ng/mL. Biochemical recurrence (BCR) was determined using the Phoenix definition. Multivariate linear regression analysis was performed to detect the related factors to achieve this nadir. RESULTS We assigned 409, 592, and 88 patients to the low-, intermediate-risk, and high-risk groups, respectively. The median followup time was 9.5 years. The median time to achieve a PSA nadir of ≤0.2 ng/mL was 44.0 (95% confidence interval: 42.3-45.7) months. The percentage of patients that achieved the nadir was 89.1%. BCR was noted in 107 (9.8%) patients. In the multivariate analysis of patients without BCR, younger age, larger prostate volume at implantation, higher initial PSA level, and monotherapy were significantly associated with longer time to achieve the PSA nadir. CONCLUSION The median time to achieve a PSA nadir of ≤0.2 ng/mL was 44.0 months. Some patients, however, may require a lengthy period of time to do so.
Collapse
|
7
|
Darwis NDM, Oike T, Kubo N, Gondhowiardjo SA, Ohno T. Characteristics of PSA Bounce after Radiotherapy for Prostate Cancer: A Meta-Analysis. Cancers (Basel) 2020; 12:cancers12082180. [PMID: 32764448 PMCID: PMC7465291 DOI: 10.3390/cancers12082180] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
The rate and characteristics of prostate-specific antigen (PSA) bounce post-radiotherapy remain unclear. To address this issue, we performed a meta-analysis. Reports of PSA bounce post-radiotherapy with a cutoff of 0.2 ng/mL were searched by using Medline and Web of Science. The primary endpoint was the occurrence rate, and the secondary endpoints were bounce characteristics such as amplitude, time to occurrence, nadir value, and time to nadir. Radiotherapy modality, age, risk classification, androgen deprivation therapy, and the follow-up period were extracted as clinical variables. Meta-analysis and univariate meta-regression were performed with random-effect modeling. Among 290 search-positive studies, 50 reports including 26,258 patients were identified. The rate of bounce was 31%; amplitude was 1.3 ng/mL; time to occurrence was 18 months; nadir value was 0.5 ng/mL; time to nadir was 33 months. Univariate meta-regression analysis showed that radiotherapy modality (29.7%), age (20.2%), and risk classification (12.2%) were the major causes of heterogeneity in the rate of bounce. This is the first meta-analysis of PSA bounce post-radiotherapy. The results are useful for post-radiotherapy surveillance of prostate cancer patients.
Collapse
Affiliation(s)
- Narisa Dewi Maulany Darwis
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (N.D.M.D.); (N.K.); (T.O.)
- Department of Radiation Oncology, Faculty of Medicine Universitas Indonesia—Dr. Cipto Mangunkusumo National General Hospital, Jl. Diponegoro No. 71, Jakarta Pusat, DKI Jakarta 10430, Indonesia;
| | - Takahiro Oike
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (N.D.M.D.); (N.K.); (T.O.)
- Gunma University Heavy Ion Medical Center, 3-39-22, Showa-Machi, Maebashi, Gunma 371-8511, Japan
- Correspondence:
| | - Nobuteru Kubo
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (N.D.M.D.); (N.K.); (T.O.)
- Gunma University Heavy Ion Medical Center, 3-39-22, Showa-Machi, Maebashi, Gunma 371-8511, Japan
| | - Soehartati A Gondhowiardjo
- Department of Radiation Oncology, Faculty of Medicine Universitas Indonesia—Dr. Cipto Mangunkusumo National General Hospital, Jl. Diponegoro No. 71, Jakarta Pusat, DKI Jakarta 10430, Indonesia;
| | - Tatsuya Ohno
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (N.D.M.D.); (N.K.); (T.O.)
- Gunma University Heavy Ion Medical Center, 3-39-22, Showa-Machi, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
8
|
Takakusagi Y, Katoh H, Kano K, Anno W, Tsuchida K, Mizoguchi N, Serizawa I, Yoshida D, Kamada T. Preliminary result of carbon-ion radiotherapy using the spot scanning method for prostate cancer. Radiat Oncol 2020; 15:127. [PMID: 32460889 PMCID: PMC7254700 DOI: 10.1186/s13014-020-01575-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 05/18/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Carbon-ion radiotherapy (CIRT) for prostate cancer was initiated at Kanagawa Cancer Center in 2015. The present study analyzed the preliminary clinical outcomes of CIRT for prostate cancer. METHODS The clinical outcomes of 253 patients with prostate cancer who were treated with CIRT delivered using the spot scanning method between December 2015 and December 2017 were retrospectively analyzed. The irradiation dose was set at 51.6 Gy (relative biological effectiveness) delivered in 12 fractions over 3 weeks. Biochemical relapse was defined using the Phoenix definition. Toxicities were assessed according to CTCAE version 4.0. RESULTS The median patient age was 70 (47-86) years. The median follow-up duration was 35.3 (4.1-52.9) months. According to the D'Amico classification system, 8, 88, and 157 patients were classified as having low, intermediate, and high risks, respectively. Androgen deprivation therapy was administered in 244 patients. The biochemical relapse-free rate in the low-, intermediate-, and high-risk groups at 3 years was 87.5, 88.0, and 97.5%, respectively (P = 0.036). Grade 2 acute urinary toxicity was observed in 12 (4.7%) patients. Grade 2 acute rectal toxicity was not observed. Grade 2 late urinary toxicity and grade 2 late rectal toxicity were observed in 17 (6.7%) and 3 patients (1.2%), respectively. Previous transurethral resection of the prostate was significantly associated with late grade 2 toxicity in univariate analysis. The predictive factor for late rectal toxicity was not detected. CONCLUSION The present study demonstrated that CIRT using the spot scanning method for prostate cancer produces favorable outcomes.
Collapse
Affiliation(s)
- Yosuke Takakusagi
- Department of Radiation Oncology, Kanagawa Cancer Center, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan
| | - Hiroyuki Katoh
- Department of Radiation Oncology, Kanagawa Cancer Center, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan.
| | - Kio Kano
- Department of Radiation Oncology, Kanagawa Cancer Center, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan
| | - Wataru Anno
- Department of Radiation Oncology, Kanagawa Cancer Center, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan
| | - Keisuke Tsuchida
- Department of Radiation Oncology, Kanagawa Cancer Center, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan
| | - Nobutaka Mizoguchi
- Department of Radiation Oncology, Kanagawa Cancer Center, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan
| | - Itsuko Serizawa
- Department of Radiation Oncology, Kanagawa Cancer Center, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan
| | - Daisaku Yoshida
- Department of Radiation Oncology, Kanagawa Cancer Center, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan
| | - Tadashi Kamada
- Department of Radiation Oncology, Kanagawa Cancer Center, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan
| |
Collapse
|
9
|
Nakai Y, Tanaka N, Asakawa I, Anai S, Miyake M, Morizawa Y, Hori S, Owari T, Fujii T, Yamaki K, Hasegawa M, Fujimoto K. Prostate-specific antigen bounce after 125I-brachytherapy for prostate cancer is a favorable prognosticator in patients who are biochemical recurrence-free at 4 years and correlates with testosterone. Jpn J Clin Oncol 2020; 50:58-65. [PMID: 31665354 DOI: 10.1093/jjco/hyz136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 08/04/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Because patients with prostate-specific antigen (PSA) bounce do not experience biochemical recurrence (BCR) until PSA bounce occurs, the period until PSA bounce ends can be considered the so-called lead-time bias. Therefore, we evaluated differences in BCR-free rate in prostate cancer patients who were BCR-free 4 years after 125I-brachytherapy alone. Furthermore, we evaluated predictors for PSA bounce and the correlation between testosterone and PSA bounce. METHODS From 2004 to 2012, 256 patients with prostate adenocarcinoma underwent 125I-brachytherapy alone. PSA and testosterone levels were monitored prior to 125I-brachytherapy, at 1, 3, 6, 12, 18, 24, 30, 36, 42, 48, 54 and 60 months after 125I-brachytherapy and yearly after 5-year follow-up. PSA bounce was defined as ≥0.2 ng/ml increase above the interval PSA nadir, followed by a decrease to nadir or below. RESULTS BCR-free rate in patients with PSA bounce (100% 7-year BCR-free rate) was significantly better (P < 0.044) than that in patients without PSA bounce (95.7% 7-year BCR-free rate) in patients who were BCR-free 4 years after 125I-brachytherapy alone (n = 223). Age was the only predictor (odds ratio: 0.93, 95% confidence interval: 0.88-0.98, P = 0.004) for PSA bounce (n = 177). The testosterone level at PSA bounce was significantly higher (P = 0.036) than that at nadir before PSA bounce (87 cases). CONCLUSIONS Patients with PSA bounce had good BCR-free rate even in patients who were BCR-free 4 years after 125I-brachytherapy alone. Testosterone levels were higher at PSA bounce; increased testosterone levels may be a cause of PSA bounce.
Collapse
Affiliation(s)
- Yasushi Nakai
- Department of Urology, Nara Medical University, Nara, Japan
| | | | - Isao Asakawa
- Department of Radiation Oncology, Nara Medical University, Nara, Japan
| | - Satoshi Anai
- Department of Urology, Nara Medical University, Nara, Japan
| | - Makito Miyake
- Department of Urology, Nara Medical University, Nara, Japan
| | | | - Shunta Hori
- Department of Urology, Nara Medical University, Nara, Japan
| | - Takuya Owari
- Department of Urology, Nara Medical University, Nara, Japan
| | - Tomomi Fujii
- Department of Diagnostic Pathology, Nara Medical University, Nara, Japan
| | - Kaori Yamaki
- Department of Radiation Oncology, Nara Medical University, Nara, Japan
| | | | | |
Collapse
|
10
|
Darwis NDM, Oike T, Kawamura H, Kawahara M, Kubo N, Sato H, Miyasaka Y, Katoh H, Ishikawa H, Matsui H, Miyazawa Y, Ito K, Suzuki K, Gondhowiardjo S, Nakano T, Ohno T. Kinetics of Prostate-Specific Antigen after Carbon Ion Radiotherapy for Prostate Cancer. Cancers (Basel) 2020; 12:cancers12030589. [PMID: 32143495 PMCID: PMC7139713 DOI: 10.3390/cancers12030589] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/27/2020] [Accepted: 03/02/2020] [Indexed: 12/29/2022] Open
Abstract
This study aimed to first elucidate prostate-specific antigen (PSA) kinetics in prostate cancer patients treated with carbon ion radiotherapy (CIRT). From 2010 to 2015, 131 patients with prostate adenocarcinoma treated with CIRT (57.6 Gy relative biological effectiveness (RBE) in 16 fractions) alone were recruited. PSA was measured at 1, 2, 3, 6, 9, 12, 15, 18, 21, 24, 30, 36, 42, 48, 54, and 60 months post-CIRT. PSA bounce was defined as PSA increase over a cutoff followed by spontaneous decrease to or below the pre-bounce nadir. PSA failure was determined using the Phoenix criteria (nadir + 2.0 ng/mL). As a result, non-failure-associated temporary increase in PSA exhibited two distinct patterns, namely a classical bounce and a surge at one month. PSA bounce of ³0.2 ng/mL was observed in 55.7% of the patients. Bounce amplitude was <2.0 ng/mL in 97.6% of cases. Bounce occurred significantly earlier than PSA failure. Younger age was a significant predictor of bounce occurrence. Bounce positivity was a significant predictor of favorable 5-year PSA failure-free survival. Meanwhile, a PSA surge of ³0.2 ng/mL was observed in 67.9% of patients. Surge amplitude was significantly larger than bounce amplitude. Larger prostate volume was a significant predictor of PSA surge occurrence. PSA surge positivity did not significantly predict PSA failure. In summary, PSA bounce was distinguishable from PSA failure with regard to timing of occurrence and amplitude (earlier and lower for bounce, respectively). These data are useful for post-CIRT surveillance of prostate cancer patients.
Collapse
Affiliation(s)
- Narisa Dewi Maulany Darwis
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (N.D.M.D.); (H.K.); (N.K.); (H.S.); (Y.M.); (H.K.); (H.I.); (T.N.)
- Department of Radiation Oncology, Faculty of Medicine Universitas Indonesia – dr. Cipto Mangunkusumo Hospital, Jl. P. Diponegoro no. 71, Jakarta 10430, Indonesia;
| | - Takahiro Oike
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (N.D.M.D.); (H.K.); (N.K.); (H.S.); (Y.M.); (H.K.); (H.I.); (T.N.)
- Gunma University Heavy Ion Medical Center, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (H.M.); (K.S.)
- Correspondence:
| | - Hidemasa Kawamura
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (N.D.M.D.); (H.K.); (N.K.); (H.S.); (Y.M.); (H.K.); (H.I.); (T.N.)
- Gunma University Heavy Ion Medical Center, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (H.M.); (K.S.)
| | - Masahiro Kawahara
- Department of Radiation Oncology, Saku Central Hospital Advanced Care Center, 3400-28, Nakagomi, Saku, Nagano 385-0051, Japan;
| | - Nobuteru Kubo
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (N.D.M.D.); (H.K.); (N.K.); (H.S.); (Y.M.); (H.K.); (H.I.); (T.N.)
- Gunma University Heavy Ion Medical Center, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (H.M.); (K.S.)
| | - Hiro Sato
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (N.D.M.D.); (H.K.); (N.K.); (H.S.); (Y.M.); (H.K.); (H.I.); (T.N.)
- Gunma University Heavy Ion Medical Center, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (H.M.); (K.S.)
| | - Yuhei Miyasaka
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (N.D.M.D.); (H.K.); (N.K.); (H.S.); (Y.M.); (H.K.); (H.I.); (T.N.)
| | - Hiroyuki Katoh
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (N.D.M.D.); (H.K.); (N.K.); (H.S.); (Y.M.); (H.K.); (H.I.); (T.N.)
- Department of Radiation Oncology, Kanagawa Cancer Center, 2-3-2, Nakao, Asahi-ku, Yokohama, Kanagawa 241-8515, Japan
| | - Hitoshi Ishikawa
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (N.D.M.D.); (H.K.); (N.K.); (H.S.); (Y.M.); (H.K.); (H.I.); (T.N.)
- Department of Radiation Oncology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Hiroshi Matsui
- Gunma University Heavy Ion Medical Center, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (H.M.); (K.S.)
- Department of Urology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (Y.M.); (K.I.)
| | - Yoshiyuki Miyazawa
- Department of Urology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (Y.M.); (K.I.)
| | - Kazuto Ito
- Department of Urology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (Y.M.); (K.I.)
- Institute for Preventive Medicine, Kurosawa Hospital, 187, Yanaka-machi, Takasaki, Gunma 370-1203, Japan
| | - Kazuhiro Suzuki
- Gunma University Heavy Ion Medical Center, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (H.M.); (K.S.)
- Department of Urology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (Y.M.); (K.I.)
| | - Soehartati Gondhowiardjo
- Department of Radiation Oncology, Faculty of Medicine Universitas Indonesia – dr. Cipto Mangunkusumo Hospital, Jl. P. Diponegoro no. 71, Jakarta 10430, Indonesia;
| | - Takashi Nakano
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (N.D.M.D.); (H.K.); (N.K.); (H.S.); (Y.M.); (H.K.); (H.I.); (T.N.)
- National Institutes for Quantum and Radiological Science and Technology, 4-9-1, Anagawa, Inage-ku, Chiba, Chiba 263-8555, Japan
| | - Tatsuya Ohno
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (N.D.M.D.); (H.K.); (N.K.); (H.S.); (Y.M.); (H.K.); (H.I.); (T.N.)
- Gunma University Heavy Ion Medical Center, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; (H.M.); (K.S.)
| |
Collapse
|
11
|
Roy S, Loblaw A, Cheung P, Chu W, Chung HT, Vesprini D, Ong A, Chowdhury A, Panjwani D, Pang G, Korol R, Davidson M, Ravi A, McCurdy B, Helou J, Zhang L, Mamedov A, Deabreu A, Quon HC. Prostate-specific Antigen Bounce After Stereotactic Body Radiotherapy for Prostate Cancer: A Pooled Analysis of Four Prospective Trials. Clin Oncol (R Coll Radiol) 2019; 31:621-629. [PMID: 31126725 DOI: 10.1016/j.clon.2019.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/05/2019] [Accepted: 04/01/2019] [Indexed: 12/21/2022]
Abstract
AIMS We conducted a pooled analysis of four prospective stereotactic body radiotherapy (SBRT) trials of low- and intermediate-risk prostate cancer to evaluate the incidence of prostate-specific antigen (PSA) bounce and its correlation with the time-dose-fraction schedule. The correlation between bounce with PSA response at 4 years (nadir PSA < 0.4 ng/ml) and biochemical failure-free survival (BFFS) was also explored. MATERIALS AND METHODS The study included four treatment groups: 35 Gy/five fractions once per week (QW) (TG-1; n = 84); 40 Gy/five fractions QW (TG-2; n = 100); 40 Gy/five fractions every other day (TG-3; n = 73); and 26 Gy/two fractions QW (TG-4; n = 30). PSA bounce was defined as a rise in PSA by 0.2 ng/ml (nadir + 0.2) or 2 ng/ml (nadir + 2.0) above nadir followed by a decrease back to nadir. Patients with fewer than three follow-up PSA tests were excluded from the pooled analysis. RESULTS In total, 287 patients were included, with a median follow-up of 5.0 years. The pooled 5-year cumulative incidence of bounce by nadir + 2.0 was 8%. The 2-year cumulative incidences of PSA bounce by nadir + 0.2 were 28.9, 21, 19.6 and 16.7% (P = 0.12) and by nadir + 2.0 were 7.2, 8, 2.7 and 6.7% (P = 0.32) for TG-1 to TG-4, respectively. Multivariable analysis revealed that for nadir + 2.0, pre-treatment PSA (odds ratio 0.49; 95% confidence interval 0.26-0.97) correlated with PSA bounce. Although PSA bounce by nadir + 0.2 (odds ratio 0.10; 95% confidence interval 0.04-0.24) and nadir + 2.0 (odds ratio 0.29; 95% confidence interval 0.09-0.93) was associated with a lower probability of PSA response at 4 years, there was no association between bounce by nadir + 0.2 (hazard ratio 0.36; 95% confidence interval 0.08-1.74) or nadir + 2 (hazard ratio 1.77; 95% confidence interval 0.28-11.07) with BFFS. CONCLUSION The incidence of PSA bounce was independent of time-dose-fraction schedule for prostate SBRT. One in 13 patients experienced a bounce high enough to be misinterpreted as biochemical failure, and clinicians should avoid early salvage interventions in these patients. There was no association between PSA bounce and BFFS.
Collapse
Affiliation(s)
- S Roy
- Tom Baker Cancer Center, Department of Oncology, University of Calgary, Calgary, Canada
| | - A Loblaw
- Odette Cancer Center, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Canada
| | - P Cheung
- Odette Cancer Center, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Canada
| | - W Chu
- Odette Cancer Center, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Canada
| | - H T Chung
- Odette Cancer Center, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Canada
| | - D Vesprini
- Odette Cancer Center, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Canada
| | - A Ong
- CancerCare Manitoba, University of Manitoba, Winnipeg, Canada
| | - A Chowdhury
- CancerCare Manitoba, University of Manitoba, Winnipeg, Canada
| | | | - G Pang
- Odette Cancer Center, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Canada
| | - R Korol
- Odette Cancer Center, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Canada
| | - M Davidson
- Odette Cancer Center, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Canada
| | - A Ravi
- Odette Cancer Center, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Canada
| | - B McCurdy
- CancerCare Manitoba, University of Manitoba, Winnipeg, Canada
| | - J Helou
- Princess Margaret Cancer Center, University of Toronto, Toronto, Canada
| | - L Zhang
- Odette Cancer Center, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Canada
| | - A Mamedov
- Odette Cancer Center, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Canada
| | - A Deabreu
- Odette Cancer Center, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Canada
| | - H C Quon
- Tom Baker Cancer Center, Department of Oncology, University of Calgary, Calgary, Canada.
| |
Collapse
|
12
|
D'hulst P, Mattelaer P, Darras J, Staelens L, Pottel H, Ponette D. Low-dose-rate brachytherapy as a minimally invasive curative treatment for localised prostate cancer has excellent oncological and functional outcomes: a retrospective analysis from a single centre. Cent European J Urol 2018; 71:270-275. [PMID: 30386646 PMCID: PMC6202624 DOI: 10.5173/ceju.2018.1749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 07/25/2018] [Accepted: 08/31/2018] [Indexed: 11/22/2022] Open
Abstract
Introduction Low-dose-rate (LDR) brachytherapy is a widely used therapeutic option for localised prostate cancer. The aim of this study was to analyse the oncological and functional outcomes after 10 years of experience with brachytherapy for localised prostate cancer. Material and methods All patients who underwent brachytherapy between April 2006 and September 2017 were included for analysis. Initial prostate-specific antigen (PSA) levels, tumour stages, Gleason scores, positive biopsies, prostate volumes, dosimetric parameters, and urinary symptoms were noted. Results A total of 201 patients underwent brachytherapy between April 2006 and September 2017. Of these patients, 159 had >3 years of oncological and functional follow-up. Only these relevant patients were included in the statistical analysis. This showed a significant, persistent decline in PSA levels (p <0.0001): the mean PSA was 1.2 ng/ml after 6 months, 1.1 ng/ml after 1 year, and 0.49 ng/ml after 3 years. Only 9 patients had tumour recurrence (3 patients with Gleason score 6 and 6 patients with Gleason score 7). After receiving adequate treatment, the patients underwent oncological follow-up. Important obstructive and irritative complaints were most pronounced during the first 9 months and decreased strongly after 18 months of follow-up. Conclusions LDR brachytherapy has excellent oncological outcomes with limited functional inconveniences that are adequately treatable. Our 10 years' experience shows that brachytherapy is a safe and effective method for the treatment of low-risk localised prostate cancer.
Collapse
Affiliation(s)
- Pieter D'hulst
- Az Damiaan Oostende, Department of Urology, Oostende, Belgium
| | | | - Jochen Darras
- Az Damiaan Oostende, Department of Urology, Oostende, Belgium
| | - Lorenzo Staelens
- AZ Delta Roeselare, Department of Radiation Oncology, Roeselare, Belgium
| | - Hans Pottel
- Catholic University Leuven KULAK, Department of Biostatistics, Kortrijk, Belgium
| | | |
Collapse
|
13
|
Kubo K, Wadasaki K, Kimura T, Murakami Y, Kajiwara M, Teishima J, Matsubara A, Nagata Y. Clinical features of prostate-specific antigen bounce after 125I brachytherapy for prostate cancer. JOURNAL OF RADIATION RESEARCH 2018; 59:649-655. [PMID: 30085076 PMCID: PMC6151629 DOI: 10.1093/jrr/rry059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 05/28/2018] [Indexed: 06/08/2023]
Abstract
The aim of this study was to analyse the clinical features of prostate-specific antigen (PSA) bounce and the difference between biochemical failure and large-magnitude PSA bounce. The cases of 352 patients with prostate cancer who underwent brachytherapy were analysed. PSA bounce was defined as an increase in PSA of ≥0.2 ng/ml above an initial PSA nadir, with subsequent decline to or below that initial nadir without treatment. PSA bounce +2 was defined as an increase in PSA of ≥2.0 ng/ml above the nadir with subsequent decline to or below that initial nadir without treatment. We analysed the rates, time to onset, and predictive factors for PSA bounce and PSA bounce +2. The median follow-up period at the time of evaluation was 82 months. One hundred and seventeen patients had PSA bounce; of them, 10 had PSA bounce +2. Biochemical failure occurred in 29 patients. The median times to onset of PSA bounce, PSA bounce +2, and biochemical failure were 20, 17.5 and 51 months, respectively. Younger age at implant and larger prostate volume were significant predictive factors for PSA bounce. Age was a significant factor for PSA bounce +2, and PSA bounce +2 patients were significantly younger than biochemical failure patients. The maximum duration from the date of PSA bounce +2 to the date when PSA level decreased was 12 months. Age at implant, time to onset, and 1-year follow-up after an increase in PSA level of ≥2 ng/ml above nadir level are useful for distinguishing between biochemical failure and PSA bounce +2.
Collapse
Affiliation(s)
- Katsumaro Kubo
- Department of Radiation Oncology, Hiroshima Prefectural Hospital, 1-5-54 Ujinakanda Minami-ku Hiroshima-shi, Hiroshima, Japan
| | - Koichi Wadasaki
- Department of Radiation Oncology, Hiroshima Prefectural Hospital, 1-5-54 Ujinakanda Minami-ku Hiroshima-shi, Hiroshima, Japan
| | - Tomoki Kimura
- Department of Radiation Oncology, Hiroshima University Hospital, 1-2-3 Kasumi Minami-ku Hiroshima-shi, Hiroshima, Japan
| | - Yuji Murakami
- Department of Radiation Oncology, Hiroshima University Hospital, 1-2-3 Kasumi Minami-ku Hiroshima-shi, Hiroshima, Japan
| | - Mitsuru Kajiwara
- Department of Urology, Hiroshima Prefectural Hospital, 1-5-54 Ujinakanda Minami-ku Hiroshima-shi, Hiroshima, Japan
| | - Jun Teishima
- Department of Urology, Hiroshima University Hospital, 1-2-3 Kasumi Minami-ku Hiroshima-shi, Hiroshima, Japan
| | - Akio Matsubara
- Department of Urology, Hiroshima University Hospital, 1-2-3 Kasumi Minami-ku Hiroshima-shi, Hiroshima, Japan
| | - Yasushi Nagata
- Department of Radiation Oncology, Hiroshima University Hospital, 1-2-3 Kasumi Minami-ku Hiroshima-shi, Hiroshima, Japan
| |
Collapse
|
14
|
Nakamura K, Ikeda I, Inokuchi H, Takayama K, Inoue T, Kamba T, Ogawa O, Hiraoka M, Mizowaki T. A pilot study of highly hypofractionated intensity-modulated radiation therapy over 3 weeks for localized prostate cancer. JOURNAL OF RADIATION RESEARCH 2018; 59:656-663. [PMID: 30085048 PMCID: PMC6151631 DOI: 10.1093/jrr/rry060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 06/15/2018] [Indexed: 06/08/2023]
Abstract
The purpose of this pilot study was to evaluate the feasibility of highly hypofractionated intensity-modulated radiation therapy (IMRT) in 15 fractions over 3 weeks for treating localized prostate cancer based on prostate position-based image-guided radiation therapy. Twenty-five patients with National Comprehensive Cancer Network (NCCN) very low- to unfavorable intermediate-risk prostate cancer were enrolled in this study from April 2014 to September 2015 to receive highly hypofractionated IMRT (without intraprostatic fiducial markers) delivering 54 Gy in 15 fractions over 3 weeks. Patients with intermediate-risk disease underwent neoadjuvant androgen suppression for 4-8 months. Twenty-four patients were treated with highly hypofractionated IMRT, and one was treated with conventionally fractionated IMRT because the dose constraint of the small bowel seemed difficult to achieve during the simulation. Seventeen percent had very low- or low-risk, 42% had favorable intermediate-risk, and 42% had unfavorable intermediate-risk disease according to NCCN guidelines. The median follow-up period was 31 months (range, 24-42 months). No Grade ≥3 acute toxicity was observed, and the incidence rates of Grade 2 acute genitourinary and gastrointestinal toxicities were 21% and 4%, respectively. No Grade ≥2 late toxicity was observed. Biochemical relapse was observed in one patient at 15 months, and the biochemical relapse-free survival rate was 95.8% at 2 years. A prostate-specific antigen bounce of ≥0.4 ng/ml was observed in 11 patients (46%). The highly hypofractionated IMRT regimen is feasible in patients with localized prostate cancer and is more convenient than conventionally fractionated schedules for patients and health-care providers.
Collapse
Affiliation(s)
- Kiyonao Nakamura
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Itaru Ikeda
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Haruo Inokuchi
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Japan
- Japanese Red Cross Wakayama Medical Center, 4-20 Komatsubara-dori, Wakayama City, Wakayama, Japan
| | - Kenji Takayama
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Takahiro Inoue
- Department of Urology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Tomomi Kamba
- Department of Urology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Osamu Ogawa
- Department of Urology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Masahiro Hiraoka
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Japan
- Japanese Red Cross Wakayama Medical Center, 4-20 Komatsubara-dori, Wakayama City, Wakayama, Japan
| | - Takashi Mizowaki
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
15
|
ACR Appropriateness Criteria ® Post-treatment Follow-up Prostate Cancer. J Am Coll Radiol 2018; 15:S132-S149. [DOI: 10.1016/j.jacr.2018.03.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 03/04/2018] [Indexed: 11/23/2022]
|
16
|
Time to PSA rise differentiates the PSA bounce after HDR and LDR brachytherapy of prostate cancer. J Contemp Brachytherapy 2018; 10:1-9. [PMID: 29619050 PMCID: PMC5881590 DOI: 10.5114/jcb.2018.73786] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 12/28/2017] [Indexed: 11/17/2022] Open
Abstract
Purpose To investigate the differences in prostate-specific antigen (PSA) bounce (PB) after high-dose-rate (HDR-BT) or low-dose-rate (LDR-BT) brachytherapy alone in prostate cancer patients. Materials and methods Ninety-four patients with localized prostate cancer (T1-T2cN0), age ranged 50-81 years, were treated with brachytherapy alone between 2008 and 2010. Patients were diagnosed with adenocarcinoma, Gleason score ≤ 7. The LDR-BT total dose was 144-145 Gy, in HDR-BT - 3 fractions of 10.5 or 15 Gy. The initial PSA level (iPSA) was assessed before treatment, then PSA was rated every 3 months over the first 2 years, and every 6 months during the next 3 years. Median follow-up was 3.0 years. Results Mean iPSA was 7.8 ng/ml. In 58 cases, PSA decreased gradually without PB or biochemical failure (BF). In 24% of patients, PB was observed. In 23 cases (24%), PB was observed using 0.2 ng/ml definition; in 10 cases (11%), BF was diagnosed using nadir + 2 ng/ml definition. The HDR-BT and LDR-BT techniques were not associated with higher level of PB (26 vs. 22%, p = 0.497). Time to the first PSA rise finished with PB was significantly shorter after HDR-BT then after LDR-BT (median, 10.5 vs. 18.0 months) during follow-up. Predictors for PB were observed only after HDR-BT. Androgen deprivation therapy (ADT) and higher Gleason score decreased the risk of PB (HR = 0.11, p = 0.03; HR = 0.51, p = 0.01). The higher PSA nadir and longer time to PSA nadir increased the risk of PB (HR 3.46, p = 0.02; HR 1.04, p = 0.04). There was no predictors for PB after LDR-BT. Conclusions HDR-BT and LDR-BT for low and intermediate risk prostate cancer had similar PB rate. The PB occurred earlier after HDR-BT than after LDR-BT. ADT and higher Gleason score decreased, and higher PSA nadir and longer time to PSA nadir increased the risk of PB after HDR-BT.
Collapse
|
17
|
Romesser PB, Pei X, Shi W, Zhang Z, Kollmeier M, McBride SM, Zelefsky MJ. Prostate-Specific Antigen (PSA) Bounce After Dose-Escalated External Beam Radiation Therapy Is an Independent Predictor of PSA Recurrence, Metastasis, and Survival in Prostate Adenocarcinoma Patients. Int J Radiat Oncol Biol Phys 2017; 100:59-67. [PMID: 29254782 DOI: 10.1016/j.ijrobp.2017.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 09/01/2017] [Accepted: 09/06/2017] [Indexed: 10/18/2022]
Abstract
PURPOSE To evaluate the difference in prostate-specific antigen (PSA) recurrence-free, distant metastasis-free, overall, and cancer-specific survival between PSA bounce (PSA-B) and non-bounce patients treated with dose-escalated external beam radiation therapy (DE-EBRT). METHODS AND MATERIALS During 1990-2010, 1898 prostate adenocarcinoma patients were treated with DE-EBRT to ≥75 Gy with ≥5 years follow-up. Patients receiving neoadjuvant/concurrent androgen-deprivation therapy (n=1035) or with fewer than 4 PSA values obtained 6 months or more after post-EBRT completion (n=87) were excluded. The evaluable 776 patients were treated (median, 81.0 Gy). Prostate-specific antigen bounce was defined as a ≥0.2-ng/mL increase above the interval PSA nadir, followed by a decrease to nadir or below. Prostate-specific antigen relapse was defined as post-radiation therapy PSA nadir + 2 ng/mL. Median follow-up was 9.2 years (interquartile range, 6.9-11.3 years). RESULTS One hundred twenty-three patients (15.9%) experienced PSA-B after DE-EBRT at a median of 24.6 months (interquartile range, 16.1-38.5 months). On multivariate analysis, younger age (P=.001), lower Gleason score (P=.0003), and higher radiation therapy dose (P=.0002) independently predicted PSA-B. Prostate-specific antigen bounce was independently associated with decreased risk for PSA relapse (hazard ratio [HR] 0.53; 95% confidence interval [CI] 0.33-0.85; P=.008), distant metastatic disease (HR 0.34; 95% CI 0.12-0.94; P=.04), and all-cause mortality (HR 0.53; 95% CI 0.29-0.96; P=.04) on multivariate Cox analysis. Because all 50 prostate cancer-specific deaths in patients without PSA-B were in the non-bounce cohort, competing-risks analysis was not applicable. A nonparametric competing-risks test demonstrated that patients with PSA-B had superior cancer-specific survival compared with patients without PSA-B (P=.004). CONCLUSIONS Patients treated with dose-escalated radiation therapy for prostate adenocarcinoma who experience posttreatment PSA-B have improved PSA recurrence-free survival, distant metastasis-free survival, overall survival, and cancer-specific survival outcomes.
Collapse
Affiliation(s)
- Paul B Romesser
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Xin Pei
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Weiji Shi
- Department of Statistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zhigang Zhang
- Department of Statistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marisa Kollmeier
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sean M McBride
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael J Zelefsky
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
18
|
Paoluzzi M, Losa A, Cerboneschi V, Colosimo C, Fontana N, Mangili P, Mignogna M, Nava L, Ravaglia V. Prostate-specific antigen percentage: An early predictive tool after iodine-125 interstitial brachytherapy for prostate cancer. Brachytherapy 2017. [PMID: 28642046 DOI: 10.1016/j.brachy.2017.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND After interstitial prostate iodine-125 brachytherapy (BT), prostate-specific antigen (PSA) evolution in time could predict overall biochemical relapse, but, considering the single patient, it is influenced by the presentation PSA amount and by the prostatic volume. It is also challenging to differentiate a PSA bounce from a biochemical relapse. PURPOSE To determine the usefulness of PSA percentage (PP) defined as the rate between PSA presented by a patient at time "t" and the PSA that the same patient had presented at the time of diagnosis (t0) assumed as 100% in predicting biochemical relapse and in differentiating them from PSA Bounces. METHODS AND MATERIALS We included 721 patients from Milan S. Raffaele Turro (399) and Lucca Campo di Marte (then S. Luca) Hospital (322). The mean age of patients was 66.5 years (range, 50-79). Mean followup was 150 months (range, 24-180). For each patient, PSA was recorded before and after iodine-125 BT, and PPs were calculated. Cox regression model, relative operating characteristic curves, and Kaplan-Meier regression model were elaborated, and a cutoff of 20% was defined. RESULTS We observed that PP >20% is an independent variable highly associated with relapse risk (p < 0.0001) with a sensitivity of 79.7%, a specificity of 82%, and an hazard ratio of 12.1, since the 6 months of followup. A PSA increase above the nadir should be because of bounce (sensitivity and specificity of 81.4%, p < 0.0001) if patient had experienced at 6 months a PP <20%. CONCLUSIONS PP might represent an early and useful tool, predictive of clinical outcome in patients after BT for prostate cancer.
Collapse
Affiliation(s)
- Mauro Paoluzzi
- Operative Unit of Urology, Department of Surgery, San Luca Hospital, Lucca, Italy.
| | - Andrea Losa
- Operative Unit of Radiotherapy, Department of Oncology, San Luca Hospital, Lucca, Italy
| | - Valentina Cerboneschi
- Operative Unit of Urology, Department of Surgery, San Raffaele Turro Hospital, Milan, Italy
| | - Caterina Colosimo
- Operative Unit of Urology, Department of Surgery, San Raffaele Turro Hospital, Milan, Italy
| | - Nicola Fontana
- Operative Unit of Urology, Department of Surgery, San Luca Hospital, Lucca, Italy
| | - Paola Mangili
- Operative Unit of Medical Physics, Department of Oncology, San Raffaele Turro Hospital, Milan, Italy
| | - Marcello Mignogna
- Operative Unit of Urology, Department of Surgery, San Raffaele Turro Hospital, Milan, Italy
| | - Luciano Nava
- Operative Unit of Radiotherapy, Department of Oncology, San Luca Hospital, Lucca, Italy
| | - Valentina Ravaglia
- Operative Unit of Medical Physics, Department of Oncology, San Luca Hospital, Lucca, Italy
| |
Collapse
|
19
|
Yamamoto Y, Offord CP, Kimura G, Kuribayashi S, Takeda H, Tsuchiya S, Shimojo H, Kanno H, Bozic I, Nowak MA, Bajzer Ž, Dingli D. Tumour and immune cell dynamics explain the PSA bounce after prostate cancer brachytherapy. Br J Cancer 2017; 115:195-202. [PMID: 27404586 PMCID: PMC4947697 DOI: 10.1038/bjc.2016.171] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/02/2016] [Accepted: 05/11/2016] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Interstitial brachytherapy for localised prostate cancer may be followed by transient increases in prostate-specific antigen (PSA) that resolve without therapy. Such PSA bounces may be associated with an improved outcome but often cause alarm in the patient and physician, and have defied explanation. METHODS We developed a mathematical model to capture the interactions between the tumour, radiation and anti-tumour immune response. The model was fitted to data from a large cohort of patients treated exclusively with interstitial brachytherapy. Immunohistological analysis for T-cell infiltration within the same tumours was also performed. RESULTS Our minimal model captures well the dynamics of the tumour after therapy, and suggests that a strong anti-tumour immune response coupled with the therapeutic effect of radiation on the tumour is responsible for the PSA bounce. Patients who experience a PSA bounce had a higher density of CD3 and CD8 cells within the tumour that likely contribute to the PSA bounce and the overall better outcomes observed. CONCLUSIONS Our observations provide a novel and unifying explanation for the PSA bounce in patients with early prostate cancer and also have implications for the use of immune-based therapies in such patients to improve outcomes.
Collapse
Affiliation(s)
- Yoichiro Yamamoto
- Department of Pathology, Shinshu University School of Medicine, 3-1-1, Asahi, Matsumoto, Nagano 390-8621, Japan.,Division of Diagnostic Pathology, Nippon Medical School Hospital, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Chetan P Offord
- Department of Molecular Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Go Kimura
- Department of Urology, Nippon Medical School Hospital, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Shigehiko Kuribayashi
- Department of Radiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Hayato Takeda
- Department of Urology, Nippon Medical School Hospital, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Shinichi Tsuchiya
- Division of Diagnostic Pathology, Nippon Medical School Hospital, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.,Diagnostic Pathology, Ritsuzankai Iida Hospital, Iida, Nagano 395-0056, Japan
| | - Hisashi Shimojo
- Department of Pathology, Shinshu University School of Medicine, 3-1-1, Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Hiroyuki Kanno
- Department of Pathology, Shinshu University School of Medicine, 3-1-1, Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Ivana Bozic
- Program for Evolutionary Dynamics, Harvard University, One Brattle Square, Suite 6, Cambridge, MA 02138-3758, USA
| | - Martin A Nowak
- Program for Evolutionary Dynamics, Harvard University, One Brattle Square, Suite 6, Cambridge, MA 02138-3758, USA
| | - Željko Bajzer
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - David Dingli
- Department of Molecular Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA.,Division of Hematology, Mayo Clinic, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| |
Collapse
|
20
|
Kindts I, Stellamans K, Billiet I, Pottel H, Lambrecht A. 125I brachytherapy in younger prostate cancer patients : Outcomes in low- and intermediate-risk disease. Strahlenther Onkol 2017; 193:707-713. [PMID: 28488030 DOI: 10.1007/s00066-017-1142-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 04/17/2017] [Indexed: 11/25/2022]
Abstract
PURPOSE To evaluate local recurrence in younger men treated with low-dose-rate (LDR) 125I brachytherapy (BT) for localized prostate cancer. PATIENTS AND METHODS A total of 192 patients (≤65-years-old) were treated with LDR 125I-BT ± hormone therapy. Local failure was defined as any prostate-specific antigen (PSA) rise leading to salvage treatment or biochemical failure according to the Phoenix definition. A bounce was defined as a rise in the nadir of ≥0.2 ng/mL followed by spontaneous return. Proportions were compared using Fisher's exact tests; continuous variables using the unpaired t-test or its non-parametric equivalent. Cox proportional hazards models were applied for multivariable survival analysis. RESULTS Median follow-up was 66 months. The 5‑year local recurrence-free survival was 96.1%. Biopsy-proven local recurrence developed in 13 patients, 4 had a Phoenix-defined recurrence at the last follow-up. Androgen deprivation therapy was started in 1 patient without proven recurrence. Univariable risk factors for local recurrence were: at least 50% positive biopsies, intermediate risk, treatment with neoadjuvant hormone therapy, low preimplantation volume receiving 100% of the prescribed dose, and no bounce development. Hormone-naïve patients not attaining a PSA value <0.5 ng/mL during follow-up also had a higher risk of local recurrences. Cox regression demonstrated that the variables "at least 50% positive biopsies" and "bounce" significantly impacted local failure (hazard ratio, HR 1.02 and 11.59, respectively). A bounce developed in 70 patients (36%). Younger patients and those treated with a lower activity per volume had a higher chance of developing a bounce in the Cox model (HR 0.99 and 0.04, respectively). CONCLUSION For younger men, LDR BT is a valid primary curative treatment option in low-risk and is to consider in intermediate-risk localized prostate cancer.
Collapse
Affiliation(s)
- Isabelle Kindts
- Department of Radiation Oncology, AZ Groeninge Hospital, President Kennedylaan 4, 8500, Kortrijk, Belgium.
| | - Karin Stellamans
- Department of Radiation Oncology, AZ Groeninge Hospital, President Kennedylaan 4, 8500, Kortrijk, Belgium
| | - Ignace Billiet
- Department of Urology, AZ Groeninge Hospital, Kortrijk, Belgium
| | - Hans Pottel
- Biostatistics, Faculty of Medicine, Catholic University Leuven Kulak, Kortrijk, Belgium
| | - Antoon Lambrecht
- Department of Radiation Oncology, AZ Groeninge Hospital, President Kennedylaan 4, 8500, Kortrijk, Belgium
| |
Collapse
|
21
|
Peacock M, Martell K, Taggar A, Meyer T, Smith W, Sia M, Angyalfi S, Husain S. Institutional long-term outcomes at the first Canadian center performing intraoperatively planned low-dose-rate brachytherapy alone in low- and intermediate-risk prostate cancer. Brachytherapy 2017; 16:822-830. [PMID: 28460998 DOI: 10.1016/j.brachy.2017.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 03/20/2017] [Accepted: 03/29/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE The aim of this study is to report the long-term outcomes and toxicities from a large cohort of patients with localized prostate cancer treated with low-dose-rate intraoperatively planned brachytherapy. METHODS AND MATERIALS Prostate-specific antigen levels, urinary symptoms, and erectile function were recorded at baseline, and each followup visit was then entered into a prospective database. Urinary toxicity requiring procedural intervention was retrospectively verified using an integrated electronic medical system. A separate cross-sectional survey was performed to measure postimplant sexual function. RESULTS A total of 822 patients with low and favorable intermediate-risk prostate cancer were treated at our institution between 2003 and 2013. The Kaplan-Meier estimates for biochemical recurrence for our cohort were 95% and 87% at 5 and 10 years, respectively. Cystoscopy, transurethral resection of prostate, or dilatation was required for 7.1% of 720 patients with more than 2 years of followup. At a median followup of 3.7 years, 64.4% of patients retained adequate erectile function for intercourse, with 54% of patients who were no longer sexually active postimplant reporting social factors as the primary reason. CONCLUSIONS Our institutional experience with intraoperative low-dose-rate prostate brachytherapy yielded excellent long-term results with a low incidence of urinary and sexual toxicity.
Collapse
Affiliation(s)
- Michael Peacock
- Department of Oncology, University of Calgary, Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - Kevin Martell
- Department of Oncology, University of Calgary, Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - Amandeep Taggar
- Department of Oncology, University of Calgary, Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - Tyler Meyer
- Department of Oncology, University of Calgary, Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - Wendy Smith
- Department of Oncology, University of Calgary, Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - Michael Sia
- Department of Oncology, University of Calgary, Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - Steve Angyalfi
- Department of Oncology, University of Calgary, Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - Siraj Husain
- Department of Oncology, University of Calgary, Tom Baker Cancer Centre, Calgary, Alberta, Canada.
| |
Collapse
|
22
|
Parameters predicting for prostate specific antigen response rates at one year post low-dose-rate intraoperative prostate brachytherapy. J Contemp Brachytherapy 2017; 9:99-105. [PMID: 28533796 PMCID: PMC5437084 DOI: 10.5114/jcb.2017.67198] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 03/04/2017] [Indexed: 11/17/2022] Open
Abstract
Purpose To develop a model for prostate specific antigen (PSA) values at one year among patients treated with intraoperatively planned 125I prostate brachytherapy (IOPB). Material and methods Four hundred and deven patients treated with IOPB for prostate adenocarcinoma were divided into four groups: those with PSA values ≥ 3 ng/ml; < 3 and ≥ 2; < 2 and ≥ 1 or PSA < 1 between 10.5 and 14.5 months post implantation (1yPSA). Ordinal regression analysis was then performed between patient, tumor, and treatment characteristics. 1yPSA values were also compared with toxicity outcomes. Results Median 1yPSA was 0.77 (0.04-17.36). Thirty-two patients (8%) had a PSA ≥ 3; 35 (9%) had PSA < 3, ≥ 2; 87 (21%) had PSA < 2, ≥ 1, and most patients 254 (62%) had PSA < 1. PSA response was independent of gland volume, Gleason score, clinical stage, seed activity, V90, V200, D90, or number of needles and seeds used. Older patients had significantly lower 1yPSA; median ages 65.1 (46.5-81.0), 62.1 (50.4-79.5), 60.5 (47.1-80.3), and 58.1 (45.1-74.2) years for each of the 1yPSA groups respectively (p < 0.001). Also, both implant V150 (p < 0.001) and initial PSA values (p = 0.04) were predictive of 1yPSA values. There was no correlation between 1yPSA values and toxicity encountered. Conclusions PSA response at 1 year post IOPB appears to be dependent on patient age, initial PSA, and implant V150. Our results provide reassurance that parameters other than biochemical failure influence 1yPSA values.
Collapse
|
23
|
Davis BJ, Taira AV, Nguyen PL, Assimos DG, D'Amico AV, Gottschalk AR, Gustafson GS, Keole SR, Liauw SL, Lloyd S, McLaughlin PW, Movsas B, Prestidge BR, Showalter TN, Vapiwala N. ACR appropriateness criteria: Permanent source brachytherapy for prostate cancer. Brachytherapy 2016; 16:266-276. [PMID: 27964905 DOI: 10.1016/j.brachy.2016.10.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 10/10/2016] [Indexed: 11/30/2022]
Abstract
PURPOSE To provide updated American College of Radiology (ACR) appropriateness criteria for transrectal ultrasound-guided transperineal interstitial permanent source brachytherapy. METHODS AND MATERIALS The ACR appropriateness criteria are evidence-based guidelines for specific clinical conditions that are reviewed every 3 years by a multidisciplinary expert panel. The guideline development and review include an extensive analysis of current medical literature from peer reviewed journals and the application of a well-established consensus methodology (modified Delphi) to rate the appropriateness of imaging and treatment procedures by the panel. In those instances where evidence is lacking or not definitive, expert opinion may be used to recommend imaging or treatment. RESULTS Permanent prostate brachytherapy (PPB) is a treatment option for appropriately selected patients with localized prostate cancer with low to very high risk disease. PPB monotherapy remains an appropriate and effective curative treatment for low-risk prostate cancer patients demonstrating excellent long-term cancer control and acceptable morbidity. PPB monotherapy can be considered for select intermediate-risk patients with multiparametric MRI useful in evaluation of such patients. High-risk patients treated with PPB should receive supplemental external beam radiotherapy (EBRT) along with androgen deprivation. Similarly, patients with involved pelvic lymph nodes may also be considered for such combined treatment but reported long-term outcomes are limited. Computed tomography-based postimplant dosimetry completed within 60 days of PPB is essential for quality assurance. PPB may be considered for treatment of local recurrence after EBRT but is associated with an increased risk of toxicity. CONCLUSIONS Updated appropriateness criteria for patient evaluation, selection, treatment, and postimplant dosimetry are given. These criteria are intended to be advisory only with the final responsibility for patient care residing with the treating clinicians.
Collapse
Affiliation(s)
- Brian J Davis
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN.
| | - Al V Taira
- Dorothy Schneider Cancer Center, San Mateo, CA
| | - Paul L Nguyen
- Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, MA
| | - Dean G Assimos
- Department of Urology, University of Alabama School of Medicine, Birmingham, AL; American Urological Association, Linthicum, MD
| | - Anthony V D'Amico
- Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, MA; American Society of Clinical Oncology, Alexandria, VA
| | - Alexander R Gottschalk
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA
| | | | - Sameer R Keole
- Department of Radiation Oncology, Mayo Clinic Scottsdale, Phoenix, AZ
| | - Stanley L Liauw
- Department of Radiation and Cellular Oncology, The University of Chicago Medical Center, Chicago, IL
| | - Shane Lloyd
- Department of Radiation Oncology, Huntsman Cancer Hospital, Salt Lake City, UT
| | | | - Benjamin Movsas
- Department of Radiation Oncology, Henry Ford Health System, Detroit, MI
| | | | - Timothy N Showalter
- Department of Radiation Oncology, University of Virginia, Charlottesville, VA
| | - Neha Vapiwala
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
24
|
Kishan AU, Wang PC, Upadhyaya SK, Hauswald H, Demanes DJ, Nickols NG, Kamrava M, Sadeghi A, Kupelian PA, Steinberg ML, Prionas ND, Buyyounouski MK, King CR. SBRT and HDR brachytherapy produce lower PSA nadirs and different PSA decay patterns than conventionally fractionated IMRT in patients with low- or intermediate-risk prostate cancer. Pract Radiat Oncol 2016; 6:268-275. [DOI: 10.1016/j.prro.2015.11.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 10/30/2015] [Accepted: 11/05/2015] [Indexed: 11/16/2022]
|
25
|
Kim DN, Straka C, Cho LC, Lotan Y, Yan J, Kavanagh B, Raben D, Cooley S, Brindle J, Xie XJ, Pistenmaa D, Timmerman R. Early and multiple PSA bounces can occur following high-dose prostate stereotactic body radiation therapy: Subset analysis of a phase 1/2 trial. Pract Radiat Oncol 2016; 7:e43-e49. [PMID: 27637137 DOI: 10.1016/j.prro.2016.06.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 06/20/2016] [Accepted: 06/21/2016] [Indexed: 11/26/2022]
Abstract
PURPOSE We hypothesized that high-dose stereotactic body radiation therapy (SBRT) would lead to faster time to nadir and lower nadir values compared with conventional radiation therapy experiences. We now report prostate-specific antigen (PSA) kinetics following high-dose SBRT in patients treated with radiation alone. METHODS AND MATERIALS Ninety-one patients were enrolled on the phase 1/2 dose escalation study of SBRT for localized prostate cancer. All patients with at least 36 months of follow-up and without hormone therapy were included in this analysis (n = 47). Treatment response parameters evaluated include time to nadir, nadir value, occurrence of PSA bounces (rise of ≥0.2 ng/mL followed by a subsequent fall), magnitude of bounces, duration of bounces, and correlation of bounces with clinical outcomes. RESULTS Median follow-up was 42 months (range, 36-78 months). Treatment dose levels were 45 Gy (n = 10), 47.5 Gy (n = 8), and 50 Gy (n = 29) in 5 fractions. Biochemical control rate was 98%. Median PSA at follow-up was 0.10 ± 0.20 ng/mL. Median time to nadir was 36 ± 11 months. A total of 24/47 (51.1%) patients had ≥1 PSA bounce. Median magnitude of PSA rise during bounce was 0.50 ± 1.2 ng/mL. Median time to first bounce was 9 ± 7.0 months. Median bounce duration was 3 ± 2.3 months for the first bounce and 6 ± 5.2 months for subsequent bounces. Prostate volumes <30 mL were associated with a decreased likelihood of bounce (P = .0202), and increasing prostate volume correlated with increasingly likelihood of having ≥2 bounces (P = .027). Patients reaching PSA nadir of ≤0.1 ng/mL were less likely to experience any bounce (P = .0044). CONCLUSIONS Compared with other SBRT experiences, our study demonstrated a higher PSA bounce rate, a similar or shorter median time to bounce, and a very low nadir. Prostate volume appears correlated with bounce.
Collapse
Affiliation(s)
| | | | | | - Yair Lotan
- University of Texas Southwestern, Dallas, Texas
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Tan YG, Lau Kam On W, Huang HH, Tan Wee Kiat T. Prostate specific antigen bounce after intensity-modulated radiation therapy in an Asian population. Asian J Urol 2016; 3:59-63. [PMID: 29264166 PMCID: PMC5730797 DOI: 10.1016/j.ajur.2015.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/30/2015] [Accepted: 12/08/2015] [Indexed: 12/02/2022] Open
Abstract
Objective Serum prostate specific antigen (PSA) is commonly used to evaluate treatment response after definitive radiation therapy (RT). However, PSA levels can temporarily rise without a clear reason, termed “PSA bounce”, and often engender great anxiety for both patients and physicians. The present study aimed to determine the prevalence and factors that predict “PSA bounce” after intensity-modulated radiation therapy (IMRT), and the relevance to biochemical failure and cancer recurrence in an Asian population. Methods We retrospectively reviewed 206 patients who received IMRT for prostate cancer from 2004 to 2012 in the National Cancer Centre Singapore. These patients were followed up with regular PSA monitoring. We defined “PSA bounce” as a rise of 0.1 ng/mL, followed by two consecutive falls. Patients with biochemical failure (PSA nadir + 2 ng/mL) were further evaluated for cancer recurrence. Results Sixty-one patients (29.6%) experienced “PSA bounce”, at a median time of 16 months and lasted for 12 months. Age remained the most consistent predictor of the incidence, duration and extent of “PSA bounce”. Other contributory factors included baseline PSA, Gleason score and PSA nadir. Hormonal therapy and prostate volume did not affect this phenomenon. Sixteen patients (7.8%) developed biochemical recurrence, at median time of 32 months, of which 11 were confirmed to have metastatic disease. The median follow-up time was 71 months. Conclusion A younger age predicts PSA bounce incidence, duration and magnitude. The extent of bounce appears to be lower in Asian population. The interval to occurrence and extent of PSA elevation separates PSA bounce from disease recurrence.
Collapse
Affiliation(s)
- Yu Guang Tan
- Department of Urology, Singapore General Hospital, Singapore
- Corresponding author.
| | | | - Hong Hong Huang
- Department of Urology, Singapore General Hospital, Singapore
| | | |
Collapse
|
27
|
Al-Shraideh Y, Sejpal SV, Meeks JJ. Radiation-Resistant Prostate Cancer and Salvage Prostatectomy. Prostate Cancer 2016. [DOI: 10.1016/b978-0-12-800077-9.00031-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
28
|
Matsushima M, Kikuchi E, Matsumoto K, Kosaka T, Mizuno R, Ohashi T, Mikami S, Miyajima A, Shigematsu N, Oya M. Erectile function status is highly associated with prostate-specific antigen bounce in localized prostate cancer patients treated with permanent prostate brachytherapy. Int J Urol 2015; 23:247-52. [PMID: 26663514 DOI: 10.1111/iju.13029] [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: 05/14/2015] [Accepted: 11/09/2015] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To examine the relationship between erectile function status and prostate-specific antigen bounce after prostate brachytherapy for localized prostate cancer. METHODS We identified 154 patients who were followed up for at least 24 months after brachytherapy. Erectile function status was assessed prospectively before brachytherapy (baseline), and 3, 6, 12, 18, 24 and 36 months postoperatively using the International Index of Erectile Function-15 questionnaire. Prostate-specific antigen bounce was defined as an increase of at least 0.4 ng/mL from a previous prostate-specific antigen level with a subsequent decline equal to, or less than, the initial nadir without treatment. A logistic regression analysis was used to identify a significant set of independent predictors of prostate-specific antigen bounce after brachytherapy. RESULTS Prostate-specific antigen bounce was observed in 38 (24.7%) men. The prostate-specific antigen bounce group had a higher erectile function domain score, higher orgasmic function domain score, and higher total International Index of Erectile Function-15 score before (at baseline) and after brachytherapy (3, 6, 12, 18, 24 and 36 months after brachytherapy) than their counterparts (P < 0.05). Of the 77 patients who completed the International Index of Erectile Function-15 questionnaire 18 months after brachytherapy (the median time of prostate-specific antigen bounce), sexual desire and intercourse satisfaction domain scores, and total International Index of Erectile Function scores 18 months after brachytherapy correlated with the occurrence of prostate-specific antigen bounce. A multivariate analysis identified the intercourse satisfaction domain score 18 months after brachytherapy as an independent indicator for the occurrence of prostate-specific antigen bounce (P = 0.008). CONCLUSIONS International Index of Erectile Function-15 score seems to be correlated with the prostate-specific antigen bounce in prostate cancer patients undergoing brachytherapy, and an occurrence of prostate-specific antigen bounce seems to be more likely in those who are more sexually active.
Collapse
Affiliation(s)
| | - Eiji Kikuchi
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | | | - Takeo Kosaka
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Ryuichi Mizuno
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Toshio Ohashi
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Shuji Mikami
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Akira Miyajima
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Naoyuki Shigematsu
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
29
|
Multiparametric MRI and post implant CT-based dosimetry after prostate brachytherapy with iodine seeds: The higher the dose to the dominant index lesion, the lower the PSA bounce. Radiother Oncol 2015; 117:258-61. [DOI: 10.1016/j.radonc.2015.08.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 07/21/2015] [Accepted: 08/03/2015] [Indexed: 11/21/2022]
|
30
|
High-dose-rate brachytherapy as a monotherapy for prostate cancer—Single-institution results of the extreme fractionation regimen. Brachytherapy 2015; 14:359-65. [DOI: 10.1016/j.brachy.2015.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 12/29/2014] [Accepted: 01/05/2015] [Indexed: 11/18/2022]
|
31
|
Pontoriero A, Iatì G, Mondello S, Midili F, Siragusa C, Brogna A, Ielo I, Anastasi G, Magno C, Pergolizzi S, De Renzis C. High-Dose Robotic Stereotactic Body Radiotherapy in the Treatment of Patients With Prostate Cancer: Preliminary Results in 26 Patients. Technol Cancer Res Treat 2015; 15:179-85. [PMID: 25586517 DOI: 10.1177/1533034614566994] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Stereotactic body radiotherapy (SBRT) can emulate high dose rate brachytherapy (HDR-BRT) dose fractionation. We report our preliminary results using SBRT in monotherapy or pre-external-beam radiotherapy (EBRT) boost in patients with localized prostate cancer (LpC). The primary end point was the evaluation of both acute and late toxicities; secondary end point was the observation of prostate-specific antigen (PSA) nadir. PATIENTS AND METHODS Patients with LpC having prostate volume ≤90 cm(3) were enrolled in the present study. Patients were treated with SBRT alone or in combined modality (SBRT + EBRT). SBRT was performed using a CyberKnife System (Accuray Incorporated, Sunnyvale, California) and fiducial tracking system. RESULTS From February 2008 to July 2013, 21 patients for monotherapy (38 Gy/4 fractions) and 5 for combined modality (9.5 Gy/2 fractions plus 46 Gy/23 fractions EBRT) were enrolled. Androgen deprivation therapy (ADT) was administered in 16 of the 26 patients. The median pretreatment PSA was 9.4 (range, 4.5-14.3) ng/mL. All patients completed the planned therapy. Acute Grade 1 toxicity was observed in 18 patients, genitourinary (GU) in 12 / 26 patients, and gastrointestinal (GI) in 6 / 26 patients. Acute Grade 2 GU toxicity was reported in 1 / 26 patients, and Grade 2 GI toxicity was observed in 2 / 26 patients. The median PSA nadir was 0.15 (range, 0.02 = 1.4) ng/mL. Late toxicities were observed in 5 / 26 patients: Grade 1 GU (3 of 26), Grade 2 GU (1 of 26), and Grade 1 GI (1 of 26). Median follow-up was 21.5 (range, 8-65) months. CONCLUSIONS Our preliminary results of SBRT "simulating" HDR for LpC confirm a minimal toxicity and an optimal PSA response. The PSA nadirs appear comparable with HDR-BRT.
Collapse
Affiliation(s)
- A Pontoriero
- Department of Biomedical Sciences and Morphological and Functional Images, University of Messina, Messina, Italy
| | - G Iatì
- A.O.U. "G. Martino", Operative Unit of Radiation Oncology, Messina, Italy
| | - S Mondello
- Department of Neuroscience Messina, University of Messina, Messina, Italy
| | - F Midili
- A.O.U. "G. Martino", Operative Unit of Radiation Oncology, Messina, Italy
| | - C Siragusa
- A.O.U. "G. Martino", Operative Unit of Radiation Oncology, Messina, Italy
| | - A Brogna
- A.O.U. "G. Martino", Operative Unit of Radiation Oncology, Messina, Italy
| | - I Ielo
- A.O.U. "G. Martino", Operative Unit of Radiation Oncology, Messina, Italy
| | - G Anastasi
- A.O.U. "G. Martino", Operative Unit of Radiation Oncology, Messina, Italy
| | - C Magno
- Department of General Surgery, Oncology and Pathology, University of Messina, Messina, Italy
| | - S Pergolizzi
- Department of Biomedical Sciences and Morphological and Functional Images, University of Messina, Messina, Italy
| | - C De Renzis
- Department of Biomedical Sciences and Morphological and Functional Images, University of Messina, Messina, Italy
| |
Collapse
|
32
|
Distinguishing prostate-specific antigen bounces from biochemical failure after low-dose-rate prostate brachytherapy. J Contemp Brachytherapy 2014; 6:247-53. [PMID: 25337125 PMCID: PMC4200178 DOI: 10.5114/jcb.2014.45093] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 07/14/2014] [Accepted: 09/30/2014] [Indexed: 11/21/2022] Open
Abstract
Purpose The purpose of this study was to characterize benign prostate-specific antigen (PSA) bounces of at least 2.0 ng/mL and biochemical failure as defined by the Phoenix definition after prostate brachytherapy at our institution, and to investigate distinguishing features between three outcome groups: patients experiencing a benign PSA bounce, biochemical failure, or neither. Material and methods Five hundred and thirty consecutive men treated with low-dose-rate brachytherapy with follow-up of at least 3 years were divided into outcome groups experiencing bounce, failure, or neither. A benign bounce was defined as a rise of at least 2.0 ng/mL over the pre-rise nadir followed by a decline to 0.5 ng/mL or below, without intervention. Patient and tumor characteristics, treatment variables, and PSA kinetics were analyzed between groups. Results Thirty-two (6.0%) men experienced benign bounces and 47 (8.9%) men experienced failure. Men experiencing a bounce were younger (p = 0.01), had a higher 6-month PSA level (p = 0.03), and took longer to reach a final nadir (p < 0.01). Compared to the failure group, men with bounce had a lower pre-treatment PSA level (p = 0.01) and experienced a rise of at least 2.0 ng/mL that occurred sooner after the implant (p < 0.01) with a faster PSA doubling time (p = 0.01). Only time to PSA rise independently differentiated between bounce and failure (p < 0.01), with a benign bounce not being seen after 36 months post-treatment. Prostate-specific antigen levels during a bounce reached levels as high as 12.6 ng/mL in this cohort, and in some cases took over 5 years to decline to below 0.5 ng/mL. Conclusions Although there is substantial overlap between the features of benign PSA bounces and failure, physicians may find it useful to evaluate the timing, absolute PSA level, initial response to treatment, and rate of rise when contemplating management for a PSA rise after low-dose-rate brachytherapy.
Collapse
|
33
|
Prostate-specific antigen bounce after high-dose-rate prostate brachytherapy and hypofractionated external beam radiotherapy. Brachytherapy 2014; 13:450-5. [PMID: 25023396 DOI: 10.1016/j.brachy.2014.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 02/12/2014] [Accepted: 05/01/2014] [Indexed: 11/20/2022]
Abstract
PURPOSE To report the frequency, timing, and magnitude of prostate-specific antigen (PSA) bounce (PB) in patients who received high-dose-rate (HDR) brachytherapy (HDRB) plus hypofractionated external beam radiation therapy (HypoRT) and to assess a possible correlation between PB and biochemical failure (BF). METHODS AND MATERIALS Patients with intermediate-risk prostate cancer received 10Gy single-fraction (192)Ir HDRB followed by 50Gy in 20 daily fractions of HypoRT without androgen deprivation therapy. All patients had a minimum 2-year followup. The PB was defined as PSA elevation higher than 0.2ng/mL from previous measurement with subsequent drop to pre-bounce level. The BF was defined as PSA nadir+2ng/mL. RESULTS A total of 114 patients treated between 2001 and 2009 were eligible for analysis. At a median followup of 66 months, the PB was found in 45 (39%) patients with a median time to bounce of 16 months (range, 3-76 months). The median time to PSA normalization after a PB was 9 months (range, 2-40 months). The median magnitude of PB was 0.45ng/mL (range, 0.2-6.62). The BF occurred in 12 (10.5%) patients of whom three had a PB. Median time to BF was 52.5 months. Four patients (3.5%) in the PB group fit the criteria for BF. CONCLUSIONS The PB is common after HDRB and HypoRT and can occur up to 76 months after treatment. It can rarely fit the criteria for BF. The time to PB is shorter than the time to BF. There is a lower incidence of BF in patients with a PB. An acknowledgment of this phenomenon should be made when interpreting PSA results during followup to prevent unnecessary interventions.
Collapse
|
34
|
Multiparametric MRI for recurrent prostate cancer post radical prostatectomy and postradiation therapy. BIOMED RESEARCH INTERNATIONAL 2014; 2014:316272. [PMID: 24967355 PMCID: PMC4055489 DOI: 10.1155/2014/316272] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 04/25/2014] [Indexed: 12/30/2022]
Abstract
The clinical suspicion of local recurrence of prostate cancer (PCa) after radical prostatectomy (RP) and after radiation therapy (RT) is based on the onset of biochemical failure. The aim of this paper was to review the current role of multiparametric-MRI (mp-MRI) in the detection of locoregional recurrence. A systematic literature search using the Medline and Cochrane Library databases was performed from January 1995 up to November 2013. Bibliographies of retrieved and review articles were also examined. Only those articles reporting complete data with clinical relevance for the present review were selected. This review article is divided into two major parts: the first one considers the role of mp-MRI in the detection of PCa local recurrence after RP; the second part provides an insight about the impact of mp-MRI in the depiction of locoregional recurrence after RT (interstitial or external beam). Published data indicate an emerging role for mp-MRI in the detection and localization of locally recurrent PCa both after RP and RT which represents an information of paramount importance to perform focal salvage treatments.
Collapse
|
35
|
Balbontin FG, Moreno SA, Bley E, Chacon R, Silva A, Morgentaler A. Long-acting testosterone injections for treatment of testosterone deficiency after brachytherapy for prostate cancer. BJU Int 2014; 114:125-30. [DOI: 10.1111/bju.12668] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
| | | | - Enrique Bley
- Unidad de Urología Hospital del Salvador; Universdad de Chile; Santiago Chile
| | - Rodrigo Chacon
- Unidad de Urología Hospital del Salvador; Universdad de Chile; Santiago Chile
| | - Andres Silva
- Unidad de Urología Hospital del Salvador; Universdad de Chile; Santiago Chile
| | | |
Collapse
|
36
|
Vu CC, Haas JA, Katz AE, Witten MR. Prostate-specific antigen bounce following stereotactic body radiation therapy for prostate cancer. Front Oncol 2014; 4:8. [PMID: 24478988 PMCID: PMC3904182 DOI: 10.3389/fonc.2014.00008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 01/14/2014] [Indexed: 11/13/2022] Open
Abstract
Introduction: Prostate-specific antigen (PSA) bounce after brachytherapy has been well-documented. This phenomenon has also been identified in patients undergoing stereotactic body radiation therapy (SBRT). While the parameters that predict PSA bounce have been extensively studied in prostate brachytherapy patients, this study is the first to analyze the clinical and pathologic predictors of PSA bounce in prostate SBRT patients. Materials and Methods: Our institution has maintained a prospective database of patients undergoing SBRT for prostate cancer since 2006. Our study population includes patients between May 2006 and November 2011 who have at least 18 months of follow-up. All patients were treated using the CyberKnife treatment system. The prescription dose was 35–36.25 Gy in five fractions. Results: One hundred twenty patients were included in our study. Median PSA follow-up was 24 months (range 18–78 months). Thirty-four (28%) patients had a PSA bounce. The median time to PSA bounce was 9 months, and the median bounce size was 0.50 ng/mL. On univariate analysis, only younger age (p = 0.011) was shown to be associated with an increased incidence of PSA bounce. Other patient factors, including race, prostate size, prior treatment by hormones, and family history of prostate cancer, did not predict PSA bounces. None of the tumor characteristics studied, including Gleason score, pre-treatment PSA, T-stage, or risk classification by NCCN guidelines, were associated with increased incidence of PSA bounces. Younger age was the only statistically significant predictor of PSA bounce on multivariate analysis (OR = 0.937, p = 0.009). Conclusion: PSA bounce, which has been reported after prostate brachytherapy, is also seen in a significant percentage of patients after CyberKnife SBRT. Close observation rather than biopsy can be considered for these patients. Younger age was the only factor that predicted PSA bounce.
Collapse
Affiliation(s)
- Charles C Vu
- Division of Radiation Oncology, Winthrop-University Hospital , New York, NY , USA ; Stony Brook School of Medicine , New York, NY , USA
| | - Jonathan A Haas
- Division of Radiation Oncology, Winthrop-University Hospital , New York, NY , USA
| | - Aaron E Katz
- Department of Urology, Winthrop-University Hospital , New York, NY , USA
| | - Matthew R Witten
- Division of Radiation Oncology, Winthrop-University Hospital , New York, NY , USA
| |
Collapse
|
37
|
Salomon L, Bastide C, Beuzeboc P, Cormier L, Fromont G, Hennequin C, Mongiat-Artus P, Peyromaure M, Ploussard G, Renard-Penna R, Rozet F, Azria D, Coloby P, Molinié V, Ravery V, Rebillard X, Richaud P, Villers A, Soulié M. Recommandations en onco-urologie 2013 du CCAFU : Cancer de la prostate. Prog Urol 2013; 23 Suppl 2:S69-101. [DOI: 10.1016/s1166-7087(13)70048-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
38
|
Chira C, Taussky D, Gruszczynski N, Meissner A, Larrivée S, Carrier JF, Donath D, Delouya G. Unusually high prostate-specific antigen bounce after prostate brachytherapy: Searching for etiologic factors. Brachytherapy 2013; 12:603-7. [PMID: 23891340 DOI: 10.1016/j.brachy.2013.05.005] [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: 01/06/2013] [Revised: 05/13/2013] [Accepted: 05/24/2013] [Indexed: 11/16/2022]
Abstract
PURPOSE Determine whether fat distribution, body mass index, or clinical and dosimetric factors are associated with prostate specific antigen (PSA) bounce (PSAb) of ≥1.6 ng/mL in patients treated with permanent seed (125)I prostate brachytherapy (PB). METHODS AND MATERIALS We identified 23 patients with a PSAb of ≥1.6 ng/mL. For each patient with a bounce, at least one control with similar age (age ± 2 years, n=31) was identified. Control patients had to have no bounce (≤0.2 ng/mL) and a most recent PSA of <1 ng/mL. CT at Day 30 after PB was used to determine the volume of subcutaneous adipose tissue, visceral adipose tissue, and peri-prostatic fat. Univariate and multivariate logistic models were used to assess the association between PSAb and adipose tissue distribution and clinical and dosimetric factors. RESULTS Mean patient age was 62.3 ± 5.3 years. Mean PSAb height was 2.7 ± 0.8 ng/mL, and mean time to bounce was 9.6 ± 4 months. More than 90% of the patients reached a PSA nadir before PSAb within 12 months post-PB. Patients showing PSAb were more likely to have a T1c disease vs. T2a (odds ratio = 18.87; 95% confidence interval: 2.32-454.55; p=0.019) and a lower seed activity per cc of prostate volume (odds ratio=0.02; 95% confidence interval=0.42-2.22; p=0.026). Neither fat distribution nor body mass index was associated with PSAb (p=0.11-0.597). CONCLUSIONS Clinical and dosimetric factors play a role in PSAb of ≥1.6 ng/mL. Fat distribution is not associated with a PSAb. There is presently no satisfactory theory to explain the etiology of PSAb.
Collapse
Affiliation(s)
- Ciprian Chira
- Departement of Radiation Oncology, Centre hospitalier de l'Université de Montréal (CHUM), Hôpital Notre-Dame Hospital, Montreal, Québec, Canada; CRCHUM-Centre de recherché du Centre Hospitalier de l'Université de Montréal, Montreal, Québec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Weier C, Haffner MC, Mosbruger T, Esopi DM, Hicks J, Zheng Q, Fedor H, Isaacs WB, De Marzo AM, Nelson WG, Yegnasubramanian S. Nucleotide resolution analysis of TMPRSS2 and ERG rearrangements in prostate cancer. J Pathol 2013; 230:174-83. [PMID: 23447416 DOI: 10.1002/path.4186] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 02/08/2013] [Accepted: 02/22/2013] [Indexed: 01/01/2023]
Abstract
TMPRSS2-ERG rearrangements occur in approximately 50% of prostate cancers and therefore represent one of the most frequently observed structural rearrangements in all cancers. However, little is known about the genomic architecture of such rearrangements. We therefore designed and optimized a pipeline involving target capture of TMPRSS2 and ERG genomic sequences coupled with paired-end next-generation sequencing to resolve genomic rearrangement breakpoints in TMPRSS2 and ERG at nucleotide resolution in a large series of primary prostate cancer specimens (n = 83). This strategy showed > 90% sensitivity and specificity in identifying TMPRSS2-ERG rearrangements, and allowed identification of intra- and inter-chromosomal rearrangements involving TMPRSS2 and ERG with known and novel fusion partners. Our results indicate that rearrangement breakpoints show strong clustering in specific intronic regions of TMPRSS2 and ERG. The observed TMPRSS2-ERG rearrangements often exhibited complex chromosomal architecture associated with several intra- and inter-chromosomal rearrangements. Nucleotide resolution analysis of breakpoint junctions revealed that the majority of TMPRSS2 and ERG rearrangements (~88%) occurred at or near regions of microhomology or involved insertions of one or more base pairs. This architecture implicates non-homologous end joining (NHEJ) and microhomology-mediated end joining (MMEJ) pathways in the generation of such rearrangements. These analyses have provided important insights into the molecular mechanisms involved in generating prostate cancer-specific recurrent rearrangements.
Collapse
Affiliation(s)
- Christopher Weier
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Prostate-Specific Antigen Bounce After High-Dose-Rate Monotherapy for Prostate Cancer. Int J Radiat Oncol Biol Phys 2013; 86:729-33. [DOI: 10.1016/j.ijrobp.2013.02.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 02/21/2013] [Accepted: 02/23/2013] [Indexed: 11/18/2022]
|
41
|
Donati OF, Jung SI, Vargas HA, Gultekin DH, Zheng J, Moskowitz CS, Hricak H, Zelefsky MJ, Akin O. Multiparametric prostate MR imaging with T2-weighted, diffusion-weighted, and dynamic contrast-enhanced sequences: are all pulse sequences necessary to detect locally recurrent prostate cancer after radiation therapy? Radiology 2013; 268:440-50. [PMID: 23481164 DOI: 10.1148/radiol.13122149] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE To compare diagnostic accuracy of T2-weighted magnetic resonance (MR) imaging with that of multiparametric (MP) MR imaging combining T2-weighted imaging with diffusion-weighted (DW) MR imaging, dynamic contrast material-enhanced (DCE) MR imaging, or both in the detection of locally recurrent prostate cancer (PCa) after radiation therapy (RT). MATERIALS AND METHODS This retrospective HIPAA-compliant study was approved by the institutional review board; informed consent was waived. Fifty-three men (median age, 70 years) suspected of having post-RT recurrence of PCa underwent MP MR imaging, including DW and DCE sequences, within 6 months after biopsy. Two readers independently evaluated the likelihood of PCa with a five-point scale for T2-weighted imaging alone, T2-weighted imaging with DW imaging, T2-weighted imaging with DCE imaging, and T2-weighted imaging with DW and DCE imaging, with at least a 4-week interval between evaluations. Areas under the receiver operating characteristic curve (AUC) were calculated. Interreader agreement was assessed, and quantitative parameters (apparent diffusion coefficient [ADC], volume transfer constant [K(trans)], and rate constant [k(ep)]) were assessed at sextant- and patient-based levels with generalized estimating equations and the Wilcoxon rank sum test, respectively. RESULTS At biopsy, recurrence was present in 35 (66%) of 53 patients. In detection of recurrent PCa, T2-weighted imaging with DW imaging yielded higher AUCs (reader 1, 0.79-0.86; reader 2, 0.75-0.81) than T2-weighted imaging alone (reader 1, 0.63-0.67; reader 2, 0.46-0.49 [P ≤ .014 for all]). DCE sequences did not contribute significant incremental value to T2-weighted imaging with DW imaging (reader 1, P > .99; reader 2, P = .35). Interreader agreement was higher for combinations of MP MR imaging than for T2-weighted imaging alone (κ = 0.34-0.63 vs κ = 0.17-0.20). Medians of quantitative parameters differed significantly (P < .0001 to P = .0233) between benign tissue and PCa (ADC, 1.64 × 10(-3) mm(2)/sec vs 1.13 × 10(-3) mm(2)/sec; K(trans), 0.16 min(-1) vs 0.33 min(-1); k(ep), 0.36 min(-1) vs 0.62 min(-1)). CONCLUSION MP MR imaging has greater accuracy in the detection of recurrent PCa after RT than T2-weighted imaging alone, with no additional benefit if DCE is added to T2-weighted imaging and DW imaging.
Collapse
Affiliation(s)
- Olivio F Donati
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Lee YH, Son SH, Yoon SC, Yu M, Choi BO, Kim YS, Jang HS, Lee SN, Jang JS, Hwang TK. Stereotactic body radiotherapy for prostate cancer: a preliminary report. Asia Pac J Clin Oncol 2012; 10:e46-53. [PMID: 22994564 DOI: 10.1111/j.1743-7563.2012.01589.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2012] [Indexed: 11/30/2022]
Abstract
AIMS We report the results of a retrospective study of stereotactic body radiotherapy (SBRT) using a Cyberknife for prostate cancer. METHODS In all 29 patients were treated with hypofractionated SBRT using a Cyberknife at median 36 Gy in five fractions. All the patients were treated with a radical aim. Prostate-specific antigen (PSA) was evaluated at baseline and after radiotherapy. Acute (≤3 months) and late (>3 months) urinary and rectal toxicities were recorded according to the CTCAE version 4.0. RESULTS The median duration of follow up was 41 months. PSA values decreased in a time-dependent way. The median PSA nadir was 0.329 ng/mL, achieved after a median of 23 months' follow up. Two patients had a PSA failure according to the definition of nadir + 2 ng/mL. Eight patients (28%) had a benign PSA bounce at median 9 months after radiotherapy. CTCAE Grade 2 and 3 late urinary toxicities were reported in 3 and 3%, respectively. One patient had exacerbated urinary symptoms and received an operation. There were no severe late rectal toxicities. CONCLUSIONS The preliminary findings of our study suggest SBRT is feasible for prostate cancer treatment. Further studies with more patients and longer follow-up duration are required.
Collapse
Affiliation(s)
- Yun Hee Lee
- Departments of Radiation Oncology, the Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Guarneri A, Botticella A, Ragona R, Filippi AR, Munoz F, Casetta G, Gontero P, Tizzani A, Ricardi U. Prostate-specific antigen kinetics after I125-brachytherapy for prostate adenocarcinoma. World J Urol 2012; 31:411-5. [DOI: 10.1007/s00345-012-0932-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 08/12/2012] [Indexed: 10/28/2022] Open
|
44
|
Beriwal S, Smith RP, Houser C, Benoit RM. Prostate-specific antigen spikes with ¹³¹Cs brachytherapy. Is there a difference with other radioisotopes? Brachytherapy 2012; 11:457-9. [PMID: 22818407 DOI: 10.1016/j.brachy.2012.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 03/29/2012] [Accepted: 04/18/2012] [Indexed: 10/28/2022]
Abstract
PURPOSE There is a suggestion that a dose-rate effect exists for the prostate-specific antigen (PSA) spike after brachytherapy. ¹³¹Cs is a newer radioisotope with a half-life of 9.7 days that is being used for prostate brachytherapy. There is no published data on the PSA spike with this radioisotope and the goal of this study was to quantify PSA spikes with ¹³¹Cs and compare it with published data for other isotopes. METHODS AND MATERIALS We have been maintaining a prospective database for all patients treated with ¹³¹Cs prostate brachytherapy at our institution. We selected patients for whom followup PSA was available for at least 24 months. The PSA spike was defined as an increase of 0.2 ng/mL, followed by a decline to prespike level. RESULTS One hundred twenty-three patients had monotherapy, whereas 32 had external beam radiation therapy followed by a brachytherapy boost. Median followup was 36 months and mean numbers of PSAs obtained were 7. Forty-six (29.7%) patients had a PSA spike. The mean time and duration for the PSA spike were 12.5 and 8.8 months, respectively. The mean magnitude of increase and mean PSA value at increase were 0.63 and 1.56 ng/mL, respectively. CONCLUSIONS The incidence of a PSA spike in our series is consistent with reported numbers for other radioisotopes. The occurrence of the spike at 12.5 months appears to be at the early end of the spectrum reported for (125)I, but the duration and magnitude are similar to other radioisotopes.
Collapse
Affiliation(s)
- Sushil Beriwal
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA.
| | | | | | | |
Collapse
|
45
|
Frank SJ, Levy LB, van Vulpen M, Crook J, Sylvester J, Grimm P, Pugh TJ, Swanson DA. Outcomes after prostate brachytherapy are even better than predicted. Cancer 2012; 118:839-47. [PMID: 21751187 DOI: 10.1002/cncr.26307] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 05/04/2011] [Accepted: 05/09/2011] [Indexed: 02/03/2023]
Abstract
BACKGROUND During the first 3 years after prostate cancer treatment with radiation therapy, benign prostate-specific antigen (PSA) bounces are difficult for clinicians to distinguish from a biochemical recurrence, which can result in unnecessary interventions and erroneous predictions of outcomes. The objective of this study was to evaluate a commonly used PSA failure definition in a multinational, multi-institutional study after monotherapy with prostate brachytherapy. METHODS Participants were selected from 2919 men who underwent permanent prostate brachytherapy at the University Medical Center Utrecht, Princess Margaret Hospital, or Seattle Prostate Institute between 1998 and 2006. Inclusion required not having received androgen-deprivation therapy and having at least 30 months of follow-up. Failure was defined as any post-treatment use of hormone therapy, clinical relapse, or prostogram-defined biochemical (PSA) failure. Cases in which the nomogram predicted biochemical failure were evaluated at each institution to verify biochemical status over time and the actual clinical outcome at 5 years. RESULTS The median follow-up for the 1816 patients was 5.2 years. Concordance between the prostogram-predicted and actual outcomes, as measured by the Harrell c statistic, was 0.655 (95% confidence interval [CI], 0.536-0.774; P = .010) for the Princess Margaret group, 0.493 (95% CI, 0.259-0.648; P = .955) for the Seattle group, and 0.696 (95% CI, 0.648-0.744, P < .001) for the Utrecht group. The overall mean difference in biochemical recurrence-free survival at 5 years between actual outcomes and prostogram-defined outcomes was 9.2% (95% CI, 7.7%-10.6%). The total numbers of prostogram-defined and actual biochemical failures were 312 and 157, respectively (P = .001). CONCLUSIONS The widely used prostogram could not adequately distinguish a benign PSA bounce from a biochemical recurrence after prostate brachytherapy and could not be used to counsel patients about their predicted outcomes after treatment. The authors conclude that, to avoid unnecessary active interventions after treatment, clinicians should monitor PSA levels for at least 3 years and provide reassurance to patients that a PSA rise during this time is common and may not indicate a treatment failure.
Collapse
Affiliation(s)
- Steven J Frank
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
McBride SM, Wong DS, Dombrowski JJ, Harkins B, Tapella P, Hanscom HN, Collins SP, Kaplan ID. Hypofractionated stereotactic body radiotherapy in low-risk prostate adenocarcinoma: preliminary results of a multi-institutional phase 1 feasibility trial. Cancer 2011; 118:3681-90. [PMID: 22170628 DOI: 10.1002/cncr.26699] [Citation(s) in RCA: 160] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 10/13/2011] [Accepted: 10/13/2011] [Indexed: 01/26/2023]
Abstract
BACKGROUND Recent reports using extreme hypofractionated regimens in the treatment of low-risk prostate adenocarcinoma have been encouraging. Here, the authors report on their own multi-institutional experience with extreme hypofractionated stereotactic radiotherapy for early stage disease. METHODS In total, at 4 centers, 45 patients with National Comprehensive Cancer Network-defined, low-risk prostate adenocarcinoma were enrolled in a phase 1, multi-institutional trial of hypofractionated radiosurgery with a proprietary radiosurgical device (CyberKnife). Thirty-four patients received 7.5 grays (Gy) delivered in 5 fractions, 9 patients received 7.25 Gy delivered in 5 fractions, and 2 patients received other regimens. The variables evaluated were biochemical progression-free survival (bPFS), prostate-specific antigen (PSA) bounce, and toxicities. Health-related quality of life was evaluated using the Sexual Health Inventory for Men (SHIM), American Urological Association (AUA), and Expanded Prostate Cancer Index Composite (EPIC) questionnaires. RESULTS The median follow-up for surviving patients was 44.5 months (range, 0-62 months). The bPFS rate at 3 years was 97.7%. The median PSA declined from 4.9 ng/mL at diagnosis to 0.2 ng/mL at last follow-up, and the median percentage PSA decline at 12 months was 80%. Nine patients experienced at least 1 PSA bounce ≥0.4 ng/mL, and 4 patients experienced 2 PSA bounces. The median time to first PSA bounce was 11.6 months (range, 7.2-18.2 months), and the mean percentage PSA bounce was 1.07 ng/mL. There was 1 episode of late grade 3 urinary obstruction, and there were 2 episodes of late grade 3 proctitis. There was a significant late decline in SHIM and EPIC sexual scores and a small, late decline in the EPIC Bowel domain score. CONCLUSIONS In a select population, extreme hypofractionation with stereotactic radiosurgery was safe and effective for the treatment of low-risk prostate adenocarcinoma.
Collapse
Affiliation(s)
- Sean M McBride
- Harvard Radiation Oncology Program, Harvard University, 100 Blossom Street, 3, Boston, MA 02114, USA.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Paoluzzi M, Mignogna M, Lorenzini E, Valent F, Fontana N, Pinzi N, Repetti F, Ponchietti R. Is prostate-specific antigen percentage decrease predictive of clinical outcome after permanent iodine-125 interstitial brachytherapy for prostate cancer? Brachytherapy 2011; 11:277-83. [PMID: 22137870 DOI: 10.1016/j.brachy.2011.08.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 07/14/2011] [Accepted: 08/17/2011] [Indexed: 10/14/2022]
Abstract
OBJECTIVE To determine the usefulness of prostate-specific antigen (PSA) percentage (vs. pretreatment value assumed as 100%) in prediction of biochemical relapse, after iodine-125 ((125)I) permanent brachytherapy for prostate cancer, to employ a parameter independent by the initial PSA amount and by the individual prostatic volume. METHODS AND MATERIALS Our study included 133 patients, 102 still disease free (Group A) and 31 who experienced proven biochemical recurrence (Group B). PSA levels before and after (125)I brachytherapy were recorded, and PSA percentage vs. pretreatment values were calculated. Cox regression model, receiver operating characteristic curves, and Kaplan-Meier regression model with log-rank test were calculated. RESULTS We observed that, in patients submitted to brachytherapy for prostate cancer, a PSA percentage >20% of pretreatment value is highly associated with relapse risk (p<0.0001) and that this association is strongly present since t=6 months of followup (p<0.0001), with a hazard ratio near to five times (4.965), a sensitivity of 72.4%, and specificity of 79.8% related to the chosen cutoff. DISCUSSION Despite the amount of PSA is the only parameter that the clinicians can deploy to monitor patient's followup after permanent interstitial brachytherapy for prostate cancer, its evolution in time seems unable to predict early biochemical relapse as it is influenced by prostatic volume and initial PSA amount. CONCLUSIONS Our data suggest that a PSA percentage >20% of pretreatment value at 6 months might represent an early, inexpensive, and useful predictive tool of bad outcome in patients after permanent brachytherapy.
Collapse
Affiliation(s)
- Mauro Paoluzzi
- Operative Unit of Urology, Campo di Marte Hospital, ASL 2 Lucca, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Frank SJ, Arterbery VE, Hsu ICJ, Abdel-Wahab M, Ciezki JP, Hahn NM, Mohler JL, Moran BJ, Rosenthal SA, Rossi CJ, Yamada Y, Merrick G. American College of Radiology Appropriateness Criteria permanent source brachytherapy for prostate cancer. Brachytherapy 2011; 10:357-62. [DOI: 10.1016/j.brachy.2011.01.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 01/31/2011] [Indexed: 11/16/2022]
|
49
|
Peinemann F, Grouven U, Hemkens LG, Bartel C, Borchers H, Pinkawa M, Heidenreich A, Sauerland S. Low-dose rate brachytherapy for men with localized prostate cancer. Cochrane Database Syst Rev 2011:CD008871. [PMID: 21735436 DOI: 10.1002/14651858.cd008871.pub2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Localized prostate cancer is a slow growing tumor for many years for the majority of affected men. Low-dose rate brachytherapy (LDR-BT) is short-distance radiotherapy using low-energy radioactive sources. LDR-BT has been recommended for men with low risk localized prostate cancer. OBJECTIVES To assess the benefit and harm of LDR-BT compared to radical prostatectomy (RP), external beam radiotherapy (EBRT), and no primary therapy (NPT) in men with localized prostatic cancer. SEARCH STRATEGY The Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE (from 1950), and EMBASE (from 1980) were searched in June 2010 as well as online trials registers and reference lists of reviews. SELECTION CRITERIA Randomized, controlled trials comparing LDR-BT versus RP, EBRT, and NPT in men with clinically localized prostate cancer. DATA COLLECTION AND ANALYSIS Data on study methods, participants, treatment regimens, observation period and outcomes were recorded by two reviewers independently. MAIN RESULTS We identified only one RCT (N = 200; mean follow up 68 months). This trial compared LDR-BT and RP. The risk of bias was deemed high. Primary outcomes (overall survival, cause-specific mortality, or metastatic-free survival) were not reported. Biochemical recurrence-free survival at 5 years follow up was not significantly different between LDR-BT (78/85 (91.8%)) and RP (81/89 (91.0%)); P = 0.875; relative risk 0.92 (95% CI: 0.35 to 2.42).For severe adverse events reported at 6 months follow up, results favored LDR-BT for urinary incontinence (LDR-BT 0/85 (0.0%) versus RP 16/89 (18.0%); P < 0.001; relative risk 0) and favored RP for urinary irritation (LDR-BT 68/85 (80.0%) versus RP 4/89 (4.5%); P < 0.001; relative risk 17.80, 95% CI 6.79 to 46.66). The occurrence of urinary stricture did not significantly differ between the treatment groups (LDR-BT 2/85 (2.4%) versus RP 6/89 (6.7%); P = 0.221; relative risk 0.35, 95% CI: 0.07 to 1.68). Long-term information was not available.We did not identify significant differences of mean scores between treatment groups for patient-reported outcomes function and bother as well as generic health-related quality of life. AUTHORS' CONCLUSIONS Low-dose rate brachytherapy did not reduce biochemical recurrence-free survival versus radical prostatectomy at 5 years. For short-term severe adverse events, low-dose rate brachytherapy was significantly more favorable for urinary incontinence, but radical prostatectomy was significantly more favorable for urinary irritation. Evidence is based on one RCT with high risk of bias.
Collapse
Affiliation(s)
- Frank Peinemann
- Institute for Quality and Efficiency in Health Care (IQWiG), Dillenburger Str. 27, Cologne, Germany, 51105
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Peinemann F, Grouven U, Bartel C, Sauerland S, Borchers H, Pinkawa M, Heidenreich A, Lange S. Permanent interstitial low-dose-rate brachytherapy for patients with localised prostate cancer: a systematic review of randomised and nonrandomised controlled clinical trials. Eur Urol 2011; 60:881-93. [PMID: 21763066 DOI: 10.1016/j.eururo.2011.06.044] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Accepted: 06/20/2011] [Indexed: 10/18/2022]
Abstract
CONTEXT Prostate cancer (PCa) is the most common cancer in men. Permanent interstitial low-dose-rate brachytherapy (LDR-BT) is a short-distance radiation therapy in which low-energy radioactive sources are implanted permanently into the prostate. OBJECTIVE To assess the effectiveness and safety of LDR-BT compared to treatment alternatives in men with localised PCa. EVIDENCE ACQUISITION Bibliographic databases (Medline, Embase, and the Cochrane Library) were searched from inception until June 2010 for randomised and nonrandomised controlled trials comparing LDR-BT with radical prostatectomy (RP), external-beam radiation therapy (EBRT), or no primary therapy (NPT). Primary outcome was overall survival (OS). Secondary outcomes were disease-free survival (DFS), biochemical recurrence-free survival (bRFS), physician-reported severe adverse events (SAE), and patient-reported outcomes (PRO). EVIDENCE SYNTHESIS A total of 31 studies, including 1 randomised controlled trial (RCT), were identified. Risk of bias was high for all 31 studies. OS was reported in one nonrandomised controlled study; however, these data were not interpretable because of strong residual confounding. DFS was not reported. Comparison of bRFS between treatment groups is not validated; thus, results were not interpretable. Physician-reported urogenital late toxicity grade 2 to 3 was more common in the LDR-BT group when compared to the EBRT group. With respect to PRO, better scores for sexual and urinary function as well as urinary incontinence were reported for LDR-BT compared to RP. Better scores for bowel function were reported for LDR-BT compared to EBRT. CONCLUSIONS We found a low amount of evidence in studies that exclusively compared LDR-BT with other treatment modalities. LDR-BT may have some different physician-reported SAE and patient-reported outcomes. The current evidence is insufficient to allow a definitive conclusion about OS. Randomised trials focusing on long-term survival are needed to clarify the relevance of LDR-BT in patients with localised PCa.
Collapse
Affiliation(s)
- Frank Peinemann
- IQWiG Institute for Quality and Efficiency in Health Care, Cologne, Germany.
| | | | | | | | | | | | | | | |
Collapse
|