Strouthos 2017

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Radiotherapy and Oncology xxx (2017) xxx–xxx

Contents lists available at ScienceDirect

Radiotherapy and Oncology


journal homepage: www.thegreenjournal.com

Original article

High dose rate brachytherapy as monotherapy for localised prostate


cancer q
Iosif Strouthos a,⇑,1, Nikolaos Tselis b,1, Georgios Chatzikonstantinou b, Saeed Butt c, Dimos Baltas d,e,
Dimitra Bon f, Natasa Milickovic c, Nikolaos Zamboglou b
a
Department of Radiation Oncology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg; b Department of Radiotherapy and Oncology, J. W. Goethe
University of Frankfurt, Frankfurt am Main; c Division of Medical Physics, Department of Radiation Oncology, Sana Klinikum Offenbach, Offenbach; d Division of Medical Physics,
Department of Radiation Oncology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg; e German Cancer Consortium (DKTK), Partner Site Freiburg;
and f Institute for Biostatistics and Mathematical Modeling, J. W. Goethe University of Frankfurt, Frankfurt am Main, Germany

a r t i c l e i n f o a b s t r a c t

Article history: Background and purpose: To evaluate the oncological outcome of a three-implant high dose rate (HDR)
Received 3 July 2017 brachytherapy (BRT) protocol as monotherapy for clinically localised prostate cancer.
Received in revised form 28 September Material and methods: Between February 2008 and December 2012, 450 consecutive patients with clin-
2017
ically localised prostate cancer were treated with HDR monotherapy. The cohort comprised of 198 low-,
Accepted 28 September 2017
Available online xxxx
135 intermediate- and 117 high risk patients being treated with three single-fraction implants of 11.5 Gy
delivered to an intraoperative real-time, transrectal ultrasound defined planning treatment volume up to
a total physical dose of 34.5 Gy with an interfractional interval of 21 days. Fifty-eight patients (12.8%)
Keywords:
Prostate cancer
received ADT, 32 of whom were high- and 26 intermediate-risk. Biochemical failure was defined accord-
High-dose-rate ing to the Phoenix Consensus Criteria and genitourinary/gastrointestinal toxicity evaluated using the
Interstitial brachytherapy Common Toxicity Criteria for Adverse Events version 3.0.
Iridium-192 Results: The median follow-up time was 56.3 months. The 60-month overall survival, biochemical con-
Monotherapy trol and metastasis-free-survival rates were 96.2%, 95.0% and 99.0%, respectively. Toxicity was scored
per event with late Grade 2 and 3 genitourinary adverse events of 14.2% and 0.8%, respectively. Late
Grade 2 gastrointestinal toxicity amounted 0.4% with no instances of Grade 3 or greater late adverse
events to be reported.
Conclusions: Our results confirm HDR BRT to be a safe and effective monotherapeutic treatment modality
for clinically localised prostate cancer.
Ó 2017 Elsevier B.V. All rights reserved. Radiotherapy and Oncology xxx (2017) xxx–xxx

High-dose-rate (HDR) brachytherapy (BRT) has been success- sustaining momentum due to its low morbidity [6] and excellent
fully implemented as high-precision radiotherapy modality [1] long term clinical results [7–9]. In line with these experiences,
for the safe monotherapy of localised prostate cancer [2] with bio- our most recent publication reported on HDR monotherapy for
chemical control (BC) rates comparable to radical prostatectomy localised prostate cancer including 226 patients treated with a
[3], low-dose-rate (LDR) BRT [4] and dose-escalated external beam three-implant scheme [10]. This current report updates and
radiotherapy (EBRT) [5]. In the absence of phase 3 comparative expands our previous results for this three-implant approach
efficacy data, however, the optimal management of locally- encompassing in total 450 consecutive patients.
confined prostate adenocarcinoma remains controversial with
treatment assignment being influenced mainly by physician’s bias
and patient’s preference. Against this background, quality of life Patients and methods
issues are gaining increasing importance with HDR monotherapy
Patient characteristics
Since 2002, we have treated more than 1000 patients with HDR
q
This work has been presented at the Annual Meeting of the Japanese Society of monotherapy for clinically localised prostate cancer. During this
Radiation Oncology (JASTRO), November 25-27, 2016, Kyoto, Japan.
⇑ Corresponding author at: Department of Radiation Oncology, University period, three different protocols were implemented reflecting an
Medical Center Freiburg, 79106 Freiburg, Germany. evolution aiming to improve clinical workflow and patient com-
E-mail address: iosif.strouthos@uniklinik-freiburg.de (I. Strouthos). fort. From January 2002 to February 2004, 141 patients were trea-
1
Both authors contributed equally. ted with one implant of four fractions ά 9.5 Gy. From March 2004

https://doi.org/10.1016/j.radonc.2017.09.038
0167-8140/Ó 2017 Elsevier B.V. All rights reserved.

Please cite this article in press as: Strouthos I et al. High dose rate brachytherapy as monotherapy for localised prostate cancer. Radiother Oncol (2017),
https://doi.org/10.1016/j.radonc.2017.09.038
2 HDR-Brachytherapy as monotherapy by prostate cancer

to January 2008, 351 patients received two implants, separated by Table 1


14 days, each of two fractions ά 9.5 Gy. From February 2008, our Patient and tumour characteristics.

HDR scheme consists of three single-fraction implants, each Characteristics (n = 450)


delivering 11.5 Gy, with an interfractional interval of 21 days. Median follow-up (months) 56.3 (4.4–91.7)
The current report encompasses our clinical experience with this Age at treatment (years)
three-implant approach during the time frame from February Mean 69.1
2008 to December 2012 when we treated a cohort of 456 consec- Median 70.3

utive patients. Six patients, however, have been excluded from Pre-treatment PSA (ng/ml)
analysis due to loss of follow-up thus resulting in 450 patients Mean 7.5
Median 6.6
included for data evaluation. n (%)
All patients had histologically proven adenocarcinoma of the
Stage
prostate and were staged according to the American Joint Commit- T1b-c 151 (33.6%)
tee on Cancer, 6th edition, staging guidelines [11]. Pre-treatment T2a 153 (34.0%)
staging included digital rectal examination, transrectal ultrasound T2b 80 (17.8%)
(TRUS) and, if clinically indicated, computed tomography T2c 64 (14.2%)
T3a 2 (0.4%)
(CT)/magnetic resonance imaging (MRI) and bone scintigraphy.
The Memorial Sloan Kettering group definition [12] was used to Gleason Score
6 303 (67.3%)
classify patients into risk groups. Briefly, this risk stratification
7 102 (22.7%)
system divides patients into low risk (T1c-T2a and GS  6 and >7 45 (10.0%)
PSA  10), intermediate risk (T2b and/or GS = 7 and/or PSA >
Pre-treatment PSA (ng/ml)
10–20) and high risk (T2c or PSA > 20 or GS 8–10 or 2 10 403 (89.6%)
intermediate-risk-criteria). Eligibility criteria were clinically 11–20 42 (9.3%)
organ-confined disease in the absence of lower urinary tract symp- >20 5 (1.1%)
toms requiring treatment. Patients who had previous transurethral Age at treatment (years)
resection of the prostate (TURP) were not excluded from treatment <60 54 (12.0%)
but assigned at six months after resection. High-risk patients who 60–69 160 (35.6%)
70 236 (52.4%)
were clinically diagnosed as unsuitable for prostatectomy or dose- Androgen deprivation therapy 58 (12.8%)
escalated EBRT, or who rejected prostatectomy or definitive EBRT
Risk group
were also assigned for HDR monotherapy at the discretion of the Low 198 (44.0%)
treating physician. Exclusion criteria were metastatic disease, pre- Intermediate 135 (30.0%)
vious pelvic EBRT for another malignancy or previous open surgery High 117 (26.0%)
of the prostate.
A total of 58 patients (12.8%) received androgen deprivation
therapy (ADT), 32 (55.2%) of whom were high-risk, 16 (27.6%) treatment planning including catheter reconstruction and PTV/
intermediate-risk, and ten (17.2%) low-risk. Hormonal therapy OARs contouring according to the new image set. Evaluation of
was prescribed either neoadjuvantly and continued concurrently implant conformity was based on dose-volume parameters for
with radiation or adjuvantly for an overall duration of median nine PTV coverage in compliance with OAR dose constraints (Fig. 1).
months (range, 3–14 months). The duration of ADT for the sub- Dose specification was given as the mean dose on the PTV surface.
groups of low-risk, intermediate-risk, and high-risk patients was Using three single-fraction implants separated by 21 days, the pre-
median 4 months (range, 3–6 months), 6 months (range, 6– scribed reference dose was 11.5 Gy delivered to a total physical
10 months) and 9 months (range, 9–14 months), respectively. dose of 34.5 Gy. Dosimetry assessment parameters and OAR con-
Androgen deprivation therapy was supervised by the referring straints are shown in Table 2. All implants were performed under
urologists. Patient and tumour characteristics are shown in Table 1. spinal, or if indicated, general anaesthesia. All treatments were
performed using a 192Iridium HDR afterloading system (microS-
electron–HDR, Elekta-Brachytherapy, Elekta AB, Sweden). Written
Brachytherapy protocol
informed consent was obtained from all patients. In our series,
Our solely TRUS-based clinical workflow has been described in two patients were staged with cT3a disease. The PTV in these cases
detail elsewhere [10,13]. In short, transperineal catheter implanta- was defined as the prostate capsule plus 5.0 mm in all directions
tion was performed under TRUS-guidance in high-lithotomy posi- (except for the posterior rectal margin), allowing for coverage of
tion using a perineal template. For inverse preplanning, transversal extracapsular invasion as confirmed on pre-treatment pelvic MRI.
ultrasound (US) images of the prostate, bladder, urethra and ante-
rior rectal wall were acquired in real-time using a continuous
Follow-up and statistical analysis
probe movement technique and three-dimensional (3D) volumes
were reconstructed based on 1.0 mm image distance. The planning All patients presented in our department at six weeks after
target volume (PTV) was defined as the entire prostate gland with- completion of treatment and then every three months for the first
out margins. Based on the acquired 3D anatomy, appropriate vir- year, every six months for the second year and annually thereafter.
tual catheter positions were generated using the intraoperative During these visits, gastrointestinal/genitourinary toxicities were
treatment planning system Oncentra Prostate (Oncentra Brachy, evaluated-documented and the performed PSA control values
Elekta AB, Stockholm, Sweden) and dose volume histograms recorded. Radio-oncological follow up visits were independent
(DVHs) for the PTV and the organs at risk (OARs, i.e. bladder, ure- from PSA controls performed by referring urologists based on
thra and rectum) were calculated for evaluation of the anatomy- national recommendations regarding PSA controls after treatment
based dose optimisation. As the preplanning dosimetry parameters for prostate cancer (i.e. at 3 month-intervals for the first two years,
fulfilled our clinical protocol, TRUS-guided implantation of steel every 6 months for the 3rd and 4th year and annually onwards).
catheters (200 mm length, 1.9 mm diameter) was performed at Upon request additional information were deduced from attending
previously defined positions. After completion of implantation, a urologists. For the current analysis, the patient sample was
final 3D TRUS data set was acquired for intraoperative real-time deduced from our prospectively maintained database and

Please cite this article in press as: Strouthos I et al. High dose rate brachytherapy as monotherapy for localised prostate cancer. Radiother Oncol (2017),
https://doi.org/10.1016/j.radonc.2017.09.038
I. Strouthos et al. / Radiotherapy and Oncology xxx (2017) xxx–xxx 3

(a)

(b)

Fig. 1. Intraoperative real-time TRUS-based treatment planning. (a) Three-dimensional reconstruction of the prostate, organs at risk (i.e., rectum, urethra, bladder), in situ
catheters, and the source dwell positions as calculated using the real-time treatment planning system Oncentra Prostate (Oncentra Brachy, Elekta AB, Stockholm, Sweden) for
the final treatment plan; (b) Isodose distribution after anatomy-based dose optimisation using hybrid inverse planning optimisation (HIPO) in axial view. The isodose colour
code convention is: dark red = 300% {isodose = 34.5 Gy}; brown = 250% {isodose = 28.75 Gy}; orange = 200% {isodose = 23.0 Gy}; yellow = 150% {isodose = 17.25 Gy};
green = 125% {isodose = 14.38 Gy}; turquoise = 100% {isodose = 11.5 Gy}; dark blue = 50% {isodose = 5.75 Gy}.

Table 2
Total physical prescription doses and normal tissue dose constraints (as percent of prescribed reference dose).
3 3 3
Treatment scheme PTV D10/D 0.1 cm-Rectum D10/D 0.1 cm-Bladder D10/D 0.1 cm-Urethra D90 V100 V150 BED
11.5 Gy x 3 34.5 Gy 75%/80% 75%/80% 115%/120% 100% 90% 35% 299/167 Gy

Abbreviations: BED = biologically effective dose considering a a/b-ratio for prostate cancer of 1.5/3.0 Gy with a potential doubling time of Tpot = 42 days (10);
D10-Rectum = dose delivered to 10% of the rectum; D10-Bladder = dose delivered to 10% of the bladder; D10-Urethra = dose delivered to 10% of the urethra; D30.1 cm = minimum
dose to the most exposed 0.1 cm3 of the organ; D90 = dose delivered to 90% of the PTV; V100, V150 = percent of PTV receiving 100% and 150% of the prescription dose.

consequently retrospectively analysed. Gastrointestinal (GI) and persisted six months after treatment completion. Adverse events
genitourinary (GU) toxicities were documented with reference to were scored per event and the highest value noted during
the National Cancer Institute Common Toxicity Criteria for Adverse follow-up was selected to calculate toxicity percentages. Biochem-
Events, version 3.0. Follow-up ended during October 2014 and June ical relapse was defined using the Phoenix criteria (sustained post-
2015 with all patients receiving between March and June 2015 treatment PSA value > nadir +2 ng/ml) [14]. The definition of
additionally a final questionnaire assessing their current PSA level potency was noted as the ability of achieving an erection sufficient
and the presence as well as grade of adverse events at that period for intercourse. Using the Kaplan–Meier method, the likelihood of
of time. As late toxicity were referred side effects that began or events was deduced and thereafter compared using the log-rank

Please cite this article in press as: Strouthos I et al. High dose rate brachytherapy as monotherapy for localised prostate cancer. Radiother Oncol (2017),
https://doi.org/10.1016/j.radonc.2017.09.038
4 HDR-Brachytherapy as monotherapy by prostate cancer

test. A two-sided p-value of 0.05 was set as statistically signifi- scribed reference dose. The median values for V100, V150 and V200
cant. The Cox proportional hazards model was used for multivari- (percentage of the PTV receiving 100%, 150% and 200% of the pre-
ate analysis. Statistical analyses were performed with SPSS Version scribed reference dose) were V100 = 94.1% (range 90.1–97.4),
20 (SPSS, Inc, Chicago, IL, USA) and WinSTAT (WinSTAT, R. Fitch V150 = 32.3% (range 22.7–36.1) and V200 = 9.9% (range 5.9–13.2).
Software, Bad Krozingen, Germany). The respective mean values of the reported dosimetric indices
were: D90 = 105.7% (range 100.2–112.0), V100 = 93.9% (range
Results 90.1–97.4), V150 = 31.6% (range 22.7–36.1) and V200 = 9.9% (range
5.9–13.2).
Oncological outcomes A transient increase in the PSA level of >0.1 ng/ml occurred in
39% of patients with PSA nadir ranging from 0.06 ng/ml to
In our cohort of 450 patients, 22 (4.9%) patients experienced a 2.1 ng/ml. The median bounce magnitude was 0.6 ng/ml (range
biochemical relapse and 5 (1.1%) developed distant metastases. 0.2–7.9) and the median time to bounce was 13 months (range
The estimated BC was 95.0% (95% CI 92.6–97.4) at 60-months. 6–68).
The estimated overall survival (OS) and metastasis-free survival
(MFS) were 96.2% (95% CI 94.2–98.3) and 99.0% (95% CI 98.8–
100.0) at 60-months, respectively. Fig. 2(a) depicts BC, MFS and Discussion
OS of the patient’s sample.
Actuarial BC at 60 months in regard to risk group stratification Clinical data suggest that dose escalation for definitive radio-
was 96.1% (95% CI 92.4–99.3), 96.1% (95% CI 92.1–99.9), and therapy (RT) of localised prostate cancer improves biochemical
92.1% (95% CI 86.3–97.8) for low-, intermediate- and high-risk failure-free survival [15–17] thus implying that an additional pos-
patients, respectively, proven to be statistically insignificant itive impact on the therapeutic ratio may be achieved by escalating
(p = 0.5076, Fig. 2(b)). Actuarial BC at 60 months based on pre- the treatment dose while enhancing conformity. In this regard,
treatment PSA was 96.2% (95% CI 93.8–98.4) vs. 87.9% (95% CI recent research supports a low alpha/beta (a/b)-ratio for prostate
78.5–97.2) for iPSA  10 vs. > 10 ng/ml, respectively, showing sta- adenocarcinoma, similar to or lower than that of late responding
tistical significance (p = 0.0157). With regard to clinical tumour normal tissue of bladder or rectum, indicating that a hypofraction-
stage (cT), BC was 94.4% (95% CI 91.3–97.6), 96.6% (95% CI 91.9– ated dose regimen is favoured for tumour control without increas-
100.0) and 96.1% (95% CI 90.7–100.0) for patients with cT  T2a, ing late sequelae [3,18–20]. High-dose-rate BRT meets this
T2b and T2c, respectively, without statistically significant differ- objective optimally by exploiting the radiobiological advantage of
ence (p = 0.7706). At 60 months, the actuarial BC according to Glea- hypofractionation while ensuring superior 3D dosimetry. By now,
son score (GS) was 96.4% (95% CI 92.7–98.9), 94.0% (95% CI 88.7– a growing body of literature corroborates HDR as safe and effective
99.4) and 87.3% (95% CI 75.4–99.3) for patients with GS  6, 7a monotherapy with consistent intermediate- and long-term BC
and 7b, respectively, also not showing statistical significance rates over a range of risk groups [2,7–10,21–30].
(p = 0.1743). Hauswald et al. [7] reported on 448 patients with low- and
Characteristics used for multiple regression analyses to corre- intermediate-risk disease treated to a median of 43.5 Gy with 6
late with BC were cT (T2a, T2b, T2c), GS (6, 7a, 7b), iPSA fractions in 2 implants spaced 1 week apart. Of those, 42 (9%) were
(10, >10 ng/ml) and ADT. The multivariate Cox regression analy- treated with temporary ADT. At a median follow-up of 6.5 years
ses could not identify independent prognostic factors for biochem- the actuarial 6-and 10-year overall BC rate was 98.6% and 97.8%,
ical failure. The median follow-up was 56.3 months with 444, 443, respectively. No significant difference was noted between the
440 and 433 patients reaching the 24, 36, 48 and 60 month sur- low- and intermediate-risk groups with 98.9% versus 95.2% bio-
vival time points, respectively. Final follow-up questionnaires were chemical progression free survival at 10 years, respectively. The
returned by 433 (96.2%) patients during July and August 2015, authors documented no late grade 3–4 GI and 4.9% late grade 3–
while 17 (3.8%) patients were not alive at the time of reporting. 4 GU toxicity. One patient (0.2%) experienced grade 4 late GU
Follow-up details are given in Table 1. sequelae developing a urethral-rectal fistula after multiple transur-
ethral resections. Yoshioka et al. [9] reported their experience on
190 patients with intermediate- and high-risk disease who were
Toxicity
treated with total physical doses ranging from 45.5 Gy in 7 frac-
Table 3 shows the results for late GU and GI adverse events tions to 54.0 Gy in 9 fractions using a single implant over 4–5 days.
analysis for the entire cohort. Four (0.8%) patients reported late Forty-four percent of intermediate- and 94% of high-risk patients
GU complications Grade 3 and a single patient developed late received additionally temporary ADT. With 19-year maximum
Grade 4 urinary incontinence/retention requiring permanent follow-up (median 7.7 years), the authors reported an 8-year BC
urostomy. Two patients (0.4%) developed late Grade 2 GI toxicity. rate of 91% for the intermediate-risk and 77% for the high-risk
No Grade 3 or higher late GI toxicities were reported. group. There was no grade 4 toxicity at all with late grade 3 GI
Erectile dysfunction Grade 3, defined as consistent inability to and GU adverse events of 1% and 2%, respectively. These data cor-
sustain an erection sufficient for sexual intercourse despite impo- roborate the experience of other groups indicating that HDR BRT is
tence agents, was reported by 29 (6.4%) patients prior to treatment. applicable for intermediate- and selected high-risk cases
Of the 421 patients who reported Grade 0–2 erectile dysfunction [2,7,9,28,31,32]. Hoskin et al. [2] treated 197 patients within a
prior to treatment, 67 (14.9%) confirmed a decline to Grade 3 after dose-finding trial consisting predominantly of intermediate-
BRT. In the final questionnaire 354 patients (78.7%) reported an (52%) and high-risk (44%) cases. The prescribed total physical doses
erection sufficient for sexual intercourse achieved with or without were 34 Gy in 4 fractions, 36 Gy in 4 fractions, 31.5 Gy in 3 frac-
the use of erectile aids. tions, or 26 Gy in 2 fractions. The authors reported a 4-year BC rate
of 87% for the high-risk group which included clinical stages T3 in
21%, GS  8 in 10%, and PSA > 20 ng/ml in 25% of patients with 92%
Dosimetry and PSA kinetics
of cases receiving temporary ADT. The incidence of acute grade 3
Dosimetry quality assessment parameters were determined by GU morbidity was 3–7% with no cases of acute GI toxicity. Late
the DVH of the PTV. The median planned D90 value (dose delivered grade 3 GU toxicity was 3–16% with late grade 3 GI toxicity docu-
to 90% of the PTV) was 105.8% (range 100.2–112.0) of the pre- mented in 1% of patients. Our current data report a 5-year BC, MFS

Please cite this article in press as: Strouthos I et al. High dose rate brachytherapy as monotherapy for localised prostate cancer. Radiother Oncol (2017),
https://doi.org/10.1016/j.radonc.2017.09.038
I. Strouthos et al. / Radiotherapy and Oncology xxx (2017) xxx–xxx 5

(a) 1
0.9

Proportion surviving / Control


0.8
0.7
Biochemical Control
0.6
0.5
Overall Survival
0.4
0.3
Metastasis Free Survival
0.2
0.1
0
0 12 24 36 48 60 72 84 96
Months

(b) 1
0.9
0.8
Biochemical Control

0.7
0.6
Low Risk (n=198)
0.5
Intermediate Risk (n=135)
0.4
High Risk (n=117)
0.3
0.2
0.1
0
0 12 24 36 48 60 72 84 96
Months

Fig. 2. Kaplan–Meier actuarial analysis of all 450 patients for (a) biochemical control, overall survival and metastasis-free survival; (b) biochemical control by risk group
stratification.

Table 3
Late toxicity results (n = 450).

No. of occurrences (%)


Grade
Toxicity 1 2 3 4
Genitourinary
Frequency/Urgency 148 (32.9%) 21 (4.7%) 1 (0.2%) –
Dysuria 46 (10.2%) 5 (1.1%) 1 (0.2%) 0
Incontinence 47 (10.4%) 27 (6.0%) 1 (0.2%) 1 (0.2%)
Retention 72 (16.0%) 11 (2.4%) 0 0
Erectile Dysfunction 115 (25.6%) 87 (19.3%) 84 (18.7%) –
Gastrointestinal (Rectum)
Pain 7 (1.6%) 1 (0.2%) 0 0
Mucositis/Necrosis 3 (0.7%) 1 (0.2%) 0 0
Diarrhoea 5 (1.1%) 0 0 0

Please cite this article in press as: Strouthos I et al. High dose rate brachytherapy as monotherapy for localised prostate cancer. Radiother Oncol (2017),
https://doi.org/10.1016/j.radonc.2017.09.038
6
https://doi.org/10.1016/j.radonc.2017.09.038
Please cite this article in press as: Strouthos I et al. High dose rate brachytherapy as monotherapy for localised prostate cancer. Radiother Oncol (2017),

Table 4
Oncological results of HDR monotherapy for localised prostate cancer.

Study n HDR protocol Median f/u (y) Biochemical control* BED (Gy) EQD2 (Gy)
Gy/fraction Fractions (Implants) Total
Yoshioka et al. [9] 190 6.0 Gy 8 (1 Implant) 48.0 Gy 7.6 93% IR , 81% HR at 5 years 240–270 103–116
6.0 Gy 9 (1 Implant) 54.0 Gy
6.5 Gy 7 (1 Implant) 45.5 Gy

HDR-Brachytherapy as monotherapy by prostate cancer


Hauswald et al. [7] 448 7.0–7.25 Gy 6 (2 Implants) 42–43.5 Gy 6.5 98.9% LR, 95.2% IR at 10 years 238–253 102–108
Jawad et al. [8] 494 9.5 Gy 4 (1 Implant) 38.0 Gy 4.1 98% LR, 95% IR at 5 years 270–279 115–119
12.0 Gy 2 (1–2 Implants) 24.0 Gy 92% LR, 81% IR at 5 years
13.5 Gy 2 (1–2 Implants) 27.0 Gy 100% LR,79% IR at 5 years
Prada et al. [22] 60 19.0 Gy 1 (1 Implant) 19.0 Gy 6.0 66% LR, 63% IR at 6 years 260 111
Kukiełka et al. [23] 77 15.0 Gy 3 (3 Implants) 45.0 Gy 4.7 96.7% all risk groups at 5 years 495 212
Komiya et al. [28] 51 6.5 Gy 7 (1 Implant) 45.5 1.4 94% all risk groups at 17 months 243 104
Hoskin et al. [2] 197 8.5–9.0 Gy 4 (1 Implant) 34–36.0 Gy 3.1 95% IR , 87% HR at 4 years 227–252 97–108
10.5 Gy 3 (1 Implant) 31.5 Gy
13.0 Gy 2 (1 Implant) 26.0 Gy
Rogers et al. [26] 284 6.5 Gy 6 (2 Implants) 39.0 Gy 2.7 94% IR at 5 years 208 89
Zamboglou et al. [10] 718 9.5 Gy 4 (1 Implant) 38.0 Gy 4.4 95% LR, 93% IR 93% HR at 5 years 279–299 119–128
9.5 Gy 4 (2 Implants) 38.0 Gy
11.5 Gy 3 (3 Implants) 34.5 Gy
Barkati et al. [25] 79 10–11.5 Gy 3 (1 Implant) 30–34.5 3.3 85.1% LR/IR at 5 years 230–299 99–128
Demanes et al. [21] 298 7.0 Gy 6 (2 Implants) 42.0 Gy 5.2 97% LR/IR at 5 years 238–279 102–119
9.5 Gy 4 (1 Implant) 38.0 Gy
Mark et al. [24] 301 7.5 Gy 6 (2 Implants) 45.0 Gy 8.0 88% all risk groups at 8 years 270 117
Grills et al. [30] 65 9.5 Gy 4 (1 Implant) 38.0 Gy 2.9 98% LR/IR at 3 years 279 119
Current series 450 11.5 Gy 3 (3 Implants) 34.5 Gy 4.7 96.1% LR, 96.1% IR 92.1% HR at 5 years 299 128

Abbreviations: LR = low-risk group; IR = intermediate risk group; HR = high-risk group; f/u = follow-up; y = years; BED = biologically effective dose considering an a/b-ratio for prostate cancer of 1.5, EQD2 = equieffective dose
administered in 2.0 Gy-fractions considering an a/b-ratio for prostate cancer of 1.5 Gy.
*
Biochemical failure defined by the Phoenix definition [14].
I. Strouthos et al. / Radiotherapy and Oncology xxx (2017) xxx–xxx 7

and OS of 96.1%, 100.0%, and 96.3% for low-risk, 96.1%, 100.0%, low- and intermediate-risk prostate cancer with a low side effect
94.9% for intermediate-risk and 92.1%, 96.3%, 97.9% for high-risk rate. Long-term follow-up data are also available for its safe and
patients. Late grade 3 GU toxicity was experienced by 0.8% of the effective implementation in the treatment of selected cases of loca-
population with one patient experiencing late grade 4 complica- lised high-risk disease.
tions. No late Grade 3 GI adverse events were documented. Late
Grade 2 rectal morbidity was overall 0.4% with two patients requir-
Conflict of interest notification
ing symptomatic relief due to rectal mucositis.
With focus on erectile dysfunction following BRT, potency
The authors would like to ensure that no financial support has
preservation rates of 60–90% have been documented in the recent
been received in conjunction with the generation of the current
literature [6,7,10,22,26–30,33]. Ghadjar et al. [29] reported on 36
submission and none of the authors has any personal or institu-
patients with low- and intermediate-risk prostate cancer treated
tional financial interest in materials or devices described in this
with one implant and four fractions of 9.5 Gy. Fourteen percent
paper.
of patients received concomitant ADT. At a median follow-up of
6.9 years, the crude erectile function preservation rate in patients
without ADT was 75%. In the series by Hauswald et al. [7], 315 of References
the 448 treated patients (70%) were able to have an erection suffi-
[1] Milickovic N, Tselis N, Karagiannis E, Ferentinos K, Zamboglou N. Iridium-
cient for intercourse before treatment. Sexual function data suit- knife: another knife in radiation oncology. Brachytherapy 2017.
able for evaluation were available for 225 of 315 patients at a [2] Hoskin P, Rojas A, Lowe G, et al. High-dose-rate brachytherapy alone for
median of 6 years after treatment. Ability to have intercourse, with localized prostate cancer in patients at moderate or high risk of biochemical
recurrence. Int J Radiat Oncol Biol Phys 2012;82:1376–84.
or without the use of erectile aids, was reported by 60% of patients [3] Tucker SL, Thames HD, Michalski JM, et al. Estimation of alpha/beta for late
whose median age at the final assessment was 69 years. rectal toxicity based on RTOG 94–06. Int J Radiat Oncol Biol Phys
In our series, erectile dysfunction Grade 3 was reported by 6.4% 2011;81:600–5.
[4] Westendorp H, Nuver TT, Hoekstra CJ, Moerland MA, Minken AW. Edema and
of patients prior to treatment. Of the 421 patients who reported seed displacements affect intraoperative permanent prostate brachytherapy
Grade 0–2 erectile dysfunction prior to treatment, 14.9% confirmed dosimetry. Int J Radiat Oncol Biol Phys 2016;96:197–205.
a decline to Grade 3 after BRT with overall 78.7% of patients self- [5] Kono Y, Kubota K, Aruga T, et al. Swelling of the prostate gland by permanent
brachytherapy may affect seed migration. Jpn J Clin Oncol 2010;40:1159–65.
reporting an erection sufficient for sexual intercourse in the final [6] Morton GC, Hoskin PJ. Brachytherapy: current status and future strategies –
evaluation questionnaire, with a median age of 75 years at docu- can high dose rate replace low dose rate and external beam radiotherapy? Clin
mentation time. Oncol (Royal College of Radiologists (Great Britain)) 2013;25:474–82.
[7] Hauswald H, Kamrava MR, Fallon JM, et al. High-dose-rate monotherapy for
Although BRT had a negative impact on sexual function, we
localized prostate cancer: 10-year results. Int J Radiat Oncol Biol Phys
consider the rate of erectile preservation still encouraging and con- 2016;94:667–74.
sistent with other data reported for HDR monotherapy. [8] Jawad MS, Dilworth JT, Gustafson GS, et al. Outcomes associated with 3
In our sample group, tumour stage, GS and iPSA did not attain treatment schedules of high-dose-rate brachytherapy monotherapy for
favorable-risk prostate cancer. Int J Radiat Oncol Biol Phys 2016;94:657–66.
statistical significance using Kaplan–Meier actuarial estimates of [9] Yoshioka Y, Suzuki O, Isohashi F, et al. High-dose-rate brachytherapy as
freedom for biochemical failure. Likewise in the series of Yoshioka monotherapy for intermediate- and high-risk prostate cancer: clinical results
et al. [34], Hoskin et al. [2], and Rogers et al. [26] the fore men- for a median 8-year follow-up. Int J Radiat Oncol Biol Phys 2015.
[10] Zamboglou N, Tselis N, Baltas D, et al. High-dose-rate interstitial
tioned factors have not been proven to have a prognostic relevance brachytherapy as monotherapy for clinically localized prostate cancer:
in regard to biochemical failure. In our cohort, the relatively small treatment evolution and mature results. Int J Radiat Oncol Biol Phys
number of patients (12.8%) exposed to ADT, allowed for no valu- 2013;85:672–8.
[11] Greene FL, Page DL, Fleming ID, et al. AJCC Cancer Staging Manual. sixth
able statistical outcome to be extrapolated, regarding the useful ed. Philadelphia: Lippincott Raven; 2002.
impact of temporary hormonal therapy. In fact, the issue remains [12] Zelefsky MJ, Leibel SA, Gaudin PB, et al. Dose escalation with three-
unclear, with ongoing discussion in lack of definitive evidence of dimensional conformal radiation therapy affects the outcome in prostate
cancer. Int J Radiat Oncol Biol Phys 1998;41:491–500.
additional ADT within a HDR monotherapy scheme [35,36]. [13] Milickovic N, Mavroidis P, Tselis N, et al. 4D analysis of influence of patient
Overall, the clinical outcome data of HDR monotherapy reflect movement and anatomy alteration on the quality of 3D U/S-based prostate
current radiobiological considerations for optimal tumour control HDR brachytherapy treatment delivery. Med Phys 2011;38:4982–93.
[14] Roach M, Hanks G, Thames H, et al. Defining biochemical failure following
through hypofractionation. The biologically effective dose (BED)
radiotherapy with or without hormonal therapy in men with clinically
values in Table 4 range from 208 to 299 Gy with a median value localized prostate cancer: recommendations of the RTOG-ASTRO Phoenix
of 256 Gy, based on an a/b-ratio of 1.5 Gy. By employing our Consensus Conference. Int J Radiat Oncol Biol Phys 2006;65:965–74.
HDR dose-fractionation scheme, a tumour BED of 299 Gy or [15] Kuban DA, Tucker SL, Dong L, et al. Long-term results of the M. D. Anderson
randomized dose-escalation trial for prostate cancer. Int J Radiat Oncol Biol
EQD2 of 128 Gy could be generated, tendering such dose coverage Phys 2008;70:67–74.
impossible to be achieved with current EBRT techniques. However, [16] Zelefsky MJ, Yamada Y, Fuks Z, et al. Long-term results of conformal
there are some limitations in our study, including the inherent radiotherapy for prostate cancer: impact of dose escalation on biochemical
tumor control and distant metastases-free survival outcomes. Int J Radiat
drawbacks of retrospective analyses. Our data were generated Oncol Biol Phys 2008;71:1028–33.
from a prospectively maintained database which was analysed ret- [17] Kupelian PA, Ciezki J, Reddy CA, Klein EA, Mahadevan A. Effect of increasing
rospectively. Adding to that, the relation correlating patient’s tox- radiation doses on local and distant failures in patients with localized prostate
cancer. Int J Radiat Oncol Biol Phys 2008;71:16–22.
icity outcome and dosimetry profile was not investigated in our [18] Vogelius IR, Bentzen SM. Meta-analysis of the alpha/beta ratio for prostate
analysis. Finally, despite advancements in TRUS imaging, intrapro- cancer in the presence of an overall time factor: bad news, good news, or no
static calcifications may impair image quality thus posing as a lim- news? Int J Radiat Oncol Biol Phys 2013;85:89–94.
[19] Miralbell R, Roberts SA, Zubizarreta E, Hendry JH. Dose-fractionation
iting factor for the whole procedure. Therefore we recommend that sensitivity of prostate cancer deduced from radiotherapy outcomes of 5,969
the ‘‘technical eligibility” of each patient be evaluated prior to patients in seven international institutional datasets: alpha/beta = 1.4 (0.9-
treatment through a TRUS examination. 2.2) Gy. Int J Radiat Oncol Biol Phys 2012;82:e17–24.
[20] Nath R, Bice WS, Butler WM, et al. AAPM recommendations on dose
prescription and reporting methods for permanent interstitial brachytherapy
for prostate cancer: report of Task Group 137. Med Phys 2009;36:5310–22.
Conclusions [21] Demanes DJ, Martinez AA, Ghilezan M, et al. High-dose-rate monotherapy:
safe and effective brachytherapy for patients with localized prostate cancer.
Int J Radiat Oncol Biol Phys 2011;81:1286–92.
The results of the present study support the notion stating that [22] Prada PJ, Cardenal J, Blanco AG, et al. High-dose-rate interstitial brachytherapy
HDR monotherapy is an excellent modality for the management of as monotherapy in one fraction for the treatment of favorable stage prostate

Please cite this article in press as: Strouthos I et al. High dose rate brachytherapy as monotherapy for localised prostate cancer. Radiother Oncol (2017),
https://doi.org/10.1016/j.radonc.2017.09.038
8 HDR-Brachytherapy as monotherapy by prostate cancer

cancer: Toxicity and long-term biochemical results. Radiother Oncol [30] Grills IS, Martinez AA, Hollander M, et al. High dose rate brachytherapy as
2016;119:411–6. prostate cancer monotherapy reduces toxicity compared to low dose rate
[23] Kukielka AM, Dabrowski T, Walasek T, Olchawa A, Kudzia R, Dybek D. High- palladium seeds. J Urol 2004;171:1098–104.
dose-rate brachytherapy as a monotherapy for prostate cancer–Single- [31] Zelefsky MJ, Cowen D, Fuks Z, et al. Long term tolerance of high dose three-
institution results of the extreme fractionation regimen. Brachytherapy dimensional conformal radiotherapy in patients with localized prostate
2015;14:359–65. carcinoma. Cancer 1999;85:2460–8.
[24] Mark RJ, Anderson PJ, Akins RS, Nair M. Interstitial high-dose-rate [32] Tselis N, Tunn UW, Chatzikonstantinou G, et al. High dose rate brachytherapy
brachytherapy as monotherapy for early stage prostate cancer. Median 8- as monotherapy for localised prostate cancer: a hypofractionated two-implant
year results in 301 patients. Brachytherapy 2010;9:S76. approach in 351 consecutive patients. Radiat Oncol (London, England)
[25] Barkati M, Williams SG, Foroudi F, et al. High-dose-rate brachytherapy as a 2013;8:115.
monotherapy for favorable-risk prostate cancer: a Phase II trial. Int J Radiat [33] Vargas C, Ghilezan M, Hollander M, et al. A new model using number of
Oncol Biol Phys 2012;82:1889–96. needles and androgen deprivation to predict chronic urinary toxicity for high
[26] Rogers CL, Alder SC, Rogers RL, et al. High dose brachytherapy as monotherapy or low dose rate prostate brachytherapy. J Urol 2005;174:882–7.
for intermediate risk prostate cancer. J Urol 2012;187:109–16. [34] Yoshioka Y, Suzuki O, Otani Y, Yoshida K, Nose T, Ogawa K. High-dose-rate
[27] Martinez AA, Demanes J, Vargas C, Schour L, Ghilezan M, Gustafson GS. High- brachytherapy as monotherapy for prostate cancer: technique, rationale and
dose-rate prostate brachytherapy: an excellent accelerated-hypofractionated perspective. J Contem Brachyther 2014;6:91–8.
treatment for favorable prostate cancer. Am J Clin Oncol 2010;33:481–8. [35] Krauss D, Kestin L, Ye H, et al. Lack of benefit for the addition of androgen
[28] Komiya A, Fujiuchi Y, Ito T, et al. Early quality of life outcomes in patients with deprivation therapy to dose-escalated radiotherapy in the treatment of
prostate cancer managed by high-dose-rate brachytherapy as monotherapy. intermediate- and high-risk prostate cancer. Int J Radiat Oncol Biol Phys
Int J Urol 2013;20:185–92. 2011;80:1064–71.
[29] Ghadjar P, Oesch SL, Rentsch CA, et al. Late toxicity and five year outcomes [36] Demanes DJ, Ghilezan MI. High-dose-rate brachytherapy as monotherapy for
after high-dose-rate brachytherapy as a monotherapy for localized prostate prostate cancer. Brachytherapy 2014;13:529–41.
cancer. Radiat Oncol (London, England) 2014;9:122.

Please cite this article in press as: Strouthos I et al. High dose rate brachytherapy as monotherapy for localised prostate cancer. Radiother Oncol (2017),
https://doi.org/10.1016/j.radonc.2017.09.038

You might also like