SlideShare a Scribd company logo
1 of 256
Download to read offline
Salvageforradiorecurrentprostatecancer	MaxPeters2016
Salvage for radiorecurrent
prostate cancer
Max Peters
Patients undergoing primary radiotherapy for prostate cancer are at risk of recurrent
disease. Recurrences are often confined to the prostate and therefore eligible for a
second curative ablation, known as salvage. Salvage modalities are usually directed
at the entire prostate volume, due to former difficulties in assessing localised
recurrences. Because of previous radiation damage to the prostate and surrounding
organs at risk (OAR), whole-gland salvage modalities are associated with significant
toxicity rates.
As a result of recent developments in assessing prostate cancer location using multi-
parametric MRI and different biopsy techniques, the attention has shifted to a focal
treatment approach in the salvage setting. This is known as focal salvage. With this
approach only the recurrent tumour area is targeted, which seems to maintain cancer
control, but decreases toxicity rates.
Focal salvage Iodine-125 brachytherapy appears to be able to provide durable disease
control, while minimising late severe genitourinary (GU) and gastrointestinal (GI)
toxicity. However, for salvage brachytherapy in general, dose constraints are necessary
for OAR to prevent late severe complications. In the primary setting, these are
available for both late GU and GI toxicity. These restrictions are probably inaccurate
for the salvage setting, since the repair capacity of OAR is compromised by previous
radiation damage. The dose restrictions for the rectum, urethra and bladder as found
in this thesis are lower than those used for primary brachytherapy.
Before implementation of these restrictions is possible, patients should be adequately
selected for salvage. Failure rates for whole-gland salvage are often significant,
indicating a role for prediction models assessing which patients are most eligible for
treatment. Whole-gland salvage brachytherapy patients can be adequately selected
using the disease free survival interval (DFSI) after primary therapy and their PSA-
doubling time (PSADT) before salvage.
Forfocalsalvage,therearenomultivariablemodelsavailablebecauseseriesareusually
limited. The model in this thesis for focal salvage high intensity focused ultrasound
(HIFU) shows that the DFSI after primary radiotherapy, the pre-treatment PSA,
PSADT, prostate volume and T-stage as assessed on MRI can adequately predict
biochemical disease free survival up to 3 years follow-up.
Lastly, it seems focal salvage brachytherapy is the most cost-effective option,
predominantly driven by the lower treatment costs and reduced toxicity rates for
which follow-up treatment is indicated.
Salvage for radiorecurrent
prostate cancer
Max Peters
Salvage for radiorecurrent prostate cancer
PhD thesis, Utrecht University, the Netherlands
Copyright M. Peters, Utrecht, 2016
All rights reserved. No part of this publication may be reproduced, distributed, or
transmitted in any form or by any means, including photocopying, recording, or
other electronic or mechanical methods, without the prior written permission of
the publisher, except in the case of brief quotations embodied in critical reviews
and certain other noncommercial uses permitted by copyright law. Copyright of
the published or accepted articles has been transferred to the respective journals.
The research described in this thesis was performed at the department of Radiation
Oncology of the University Medical Centre Utrecht.
Cover: 	 Gold of the Azure by Joan Miró (1967), edited by Roy Sanders.
Lay out:	 Roy Sanders
Print:	 Print Support4U
ISBN:	978-90-821-9683-2
Copyright: 	
Chapter 1 Springer-Verlag GmbH, Berlin ©
2013
Chapter 3 Elsevier Ireland Ltd ©
2014
Chapter 4 Elsevier Ireland Ltd. ©
2015
Chapter 5 American Brachytherapy Society ©
2015 Epub ahead of print
Chapter 6 American Brachytherapy Society ©
2016 Accepted
Chapter 7 Elsevier Ireland Ltd. ©
2016 Accepted
Salvage for radiorecurrent
prostate cancer
Herbehandeling van prostaatkanker
recidieven na radiotherapie
(met een samenvatting in het Nederlands)
Proefschrift
ter verkrijging van de graad van doctor aan de Universiteit Utrecht
op gezag van de rector magnificus, prof.dr. G.J. van der Zwaan,
ingevolge het besluit van het college voor promoties
in het openbaar te verdedigen
op donderdag 10 maart 2016 des middags te 2.30 uur
door
Max Peters
geboren op 17 mei 1989
te Utrecht
Promotor: 	 Prof. dr. M. van Vulpen
Copromotoren: 	 Dr. J.R.N. van der Voort van Zyp		
Dr. Ir. M.A. Moerland
Dit proefschrift werd (mede) mogelijk gemaakt door financiële steun van
Varian Medical Systems, Elekta en de prostaatkankerstichting
“History repeats, but science reverberates.”
Siddhartha Mukherjee,
The Emperor of All Maladies: A Biography of Cancer
33 Chapter 1
Patterns of outcome and toxicity after salvage
prostatectomy, salvage cryosurgery and salvage Iodine-125
brachytherapy for prostate cancer recurrences after
radiation therapy: a multi-centre experience and literature
comparison.
Contents
09 Introduction and outline of the thesis
49 Chapter 2
Focal salvage therapy for local prostate cancer recurrences
after primary radiotherapy – a comprehensive review.
67 Chapter 3
Focal salvage Iodine-125 brachytherapy for prostate cancer
recurrences after primary radiotherapy: a retrospective
study regarding toxicity, biochemical outcome and quality
of life.
89 Chapter 4
Urethral and bladder dosimetry of total and focal salvage
Iodine-125 prostate brachytherapy: Late toxicity and dose
constraints.
109 Chapter 5
Rectal dose constraints for salvage Iodine-125 prostate
brachytherapy.
127 Chapter 6
Development and internal validation of a multivariable
prediction model for biochemical failure after whole-gland
salvage Iodine-125 prostate brachytherapy for recurrent
prostate cancer.
147 Chapter 7
Multivariable model development and internal validation
for prostate cancer specific survival and overall
survival after whole-gland salvage Iodine-125 prostate
brachytherapy.
167 Chapter 8
Development and internal validation of a multivariable
prediction model for biochemical failure after focal salvage
high intensity focused ultrasound for locally recurrent
prostate cancer: presentation of a risk score for individual
patient prognosis.
187 Chapter 9
Comparative cost-effectiveness of focal and total salvage
I-125 brachytherapy for recurrent prostate cancer after
primary radiotherapy.
203 Chapter 10
Discussion
Summary
233 Addenda
Samenvatting
Dankwoord
List of publications
Curriculum vitae
Introduction and
outline of the thesis
Introduction and outline
10
1. Epidemiology
In many countries, prostate cancer is the most commonly diagnosed male cancer
(excluding non-melanoma skin cancers), with an estimated 220,800 new cases and
an expected 27,540 prostate cancer related deaths in the United States in 20151
.
In the Netherlands, there were approximately 10.000 new cases in 2014. Since the
1990’s, the annual mortality rate is approximately stable at 2500 yearly2
. In 2012,
there were 1.1 million cases worldwide, with 307,000 attributable deaths3
. Prostate
cancer is often diagnosed in an early stage and these patients are usually eligible for
active surveillance or radical treatment. However, because of the high incidence,
in absolute terms it remains the cancer site with the second most cancer related
deaths in the United States (after lung cancer) and the fifth most common cause of
cancer-related death worldwide1,3
.
2. Stage migration
As opposed to many other types of cancer, there has been a dramatic rise in the
incidence of prostate cancer in the past decades, as can be seen in figure 1. Due to
several factors, a significant and increasing proportion of patients is diagnosed in
an early and often organ-confined stage. Most importantly, the dramatic rise in
incidence of localised disease is a direct consequence of the introduction of PSA-
testing in the 1990’s (figure 1)1
. Patients are screened using PSA at an earlier age
and more frequently, thereby increasing the possibility of finding indolent, organ-
confined tumours.
Figure 1: Time-trends in several tumour incidences.
Noticeable is the dramatic rise in prostate cancer
incidence1
 
Male
Prostate
Lung & bronchus
Colorectum
Urinary bladder
Thyroid Liver*Melanoma of the skin
Rateper100,000Population
0
25
50
75
100
125
150
175
200
225
250
1980 1985 1990 1995 2000 2005 2011
Introduction and outline
11
Localised disease is usually assessed using digital rectal examination (DRE) in
combination with transrectal ultrasound (TRUS)-guided biopsies. Technetium-99
scintigraphy (bone-scan) and/or CT are used to exclude metastases in high risk
localised disease (with a Gleason score ≥8 and/or PSA ≥20) or ≥cT3a tumours4
. In
addition,localiseddiseasecanbepartlypredictedbasedonnomogramsincorporating
clinical characteristics: for example Partin tables which use the clinical tumour(T)-
stage, histopathological Gleason score and the PSA-value5
.
In recent years, however, developments in diagnostic techniques have led to
increasing accuracy in assessing localised disease and partly in excluding lymph-
node or distant metastases. Multiparametric (mp) magnetic resonance imaging
(MRI) and 11C-Choline and 18F-Fluorocholine Positron-Emission Tomography in
combinationwithCT(PET-CT)havecontributedsignificantlytothisassessment4,6-14
.
Together with developments in biopsy techniques such as transperineal template
prostate mapping (TTPM) and MRI-guided biopsies, there has been a further
increaseinclassifyinglocalisedandclinicallysignificantdiseaseoverDREandTRUS-
guided biopsies11,12,15-18
. This stage migration to organ-confined and increasingly low-
risk disease has some important consequences: most importantly the potential to
curatively treat disease and thereby excluding or postponing the use of androgen
deprivation therapy (ADT) in case of metastases.
3. To treat or not to treat?
3.1 Treatment versus observation
Whether these often low-volume, low-grade tumours need radical treatment is a
challenging question. Large randomised controlled trials (RCT’s) have provided
conflicting results regarding radical treatment for early-stage prostate cancer.
The Prostate Cancer Intervention versus Observation (PIVOT)-trial found no
evidence of a survival difference in patients with localised prostate cancer who
were randomised between radical prostatectomy and watchful waiting (hazard ratio
[HR] 0.88; 95% confidence interval [CI] 0.71-1.08; p=0.22; absolute risk reduction
2.9%19
). Contrary, the Scandinavian Prostate Cancer Group Study Number 4
(SPCG-4) did find a difference in both overall and prostate cancer specific mortality,
with a number needed to treat (NNT) of approximately 8 to prevent one death20
.
However, the PIVOT-study might have been exposed to the above mentioned stage
migration, since patients were recruited mostly in the PSA-era. This was primarily
visible in the incidence of 50% non-palpable T1c-tumours. The SPGC-4 study only
had 12% T1c tumours, because patients were assessed mostly before the PSA era.
This could suggest a possible delayed effect of radical prostatectomy in the PIVOT
trial which might become visible after longer follow-up.
Introduction and outline
12
3.2 Screening
This so-called ‘lead-time bias’ also seems to be present in large screening trials for
prostate cancer, in which the ‘number needed to screen (NNS)’ and ‘number needed
to treat (NNT)’ to prevent one death due to prostate cancer seem to be decreasing
with longer follow-up. This has been observed in the European Randomised Study
of Screening for Prostate Cancer (ERSPC), in which over the years the NNS and
NNT have decreased from 1410 and 48 to 781 and 27, respectively21-23
. However,
not all studies confirm these results, with the most important example being the
US Prostate, Lung, Colorectal, and Ovarian (PLCO) cancer screening trial, which
showed no effect of annual PSA and DRE on survival. This study, however, suffered
from a significant contamination of the control group, in which 50% of the patients
underwent PSA-testing outside the study protocol24
.
A recent Cochrane meta-analysis did not show a pooled effect of PSA-screening
in five studies (including the ERSPC and PLCO) on prostate cancer specific and
overall mortality25,26
. This effect was also not present when pooling the results of
the ERSPC and PLCO separately. In addition, no effect on overall mortality has
been observed in any of the separate trials.
3.3 Side-effects of treatment
Assuming an effect of screening, the question remains whether the latest NNS of
781 and NNT of 27 from the ERSPC are worth the often severe side-effects of
primary radical treatment. Toxicity data from the PIVOT trial shows significant
incontinence rates after treatment: 17.1% in the radical prostatectomy group versus
6.3% in the observation group. Erectile dysfunction also considerably increased:
81.1% versus 44.1%. In addition, over 20% often severe 30-day surgical morbidity
was seen. Aggravation of toxicity after radical treatment is seen up to 15 years,
as assessed in a large cohort study comparing radiotherapy with prostatectomy
(n=1655). Both prostatectomy and radiotherapy showed an incontinence rate of
approximately 18% in this cohort and 90% erectile dysfunction. Radiotherapy
patients had some more bowel dysfunction at 15 years follow-up (16% versus
5.2%)27
. Whether these effects are actually related to surgery or radiotherapy will
possibly be answered by the awaited results of the ProtecT trial, an RCT evaluating
radiotherapy, radical prostatectomy and active surveillance for localised disease.
In addition, this trial is the first comparing these treatment modalities in a direct
manner. Active surveillance in the current era seems a safe option for low risk
patients, with long term excellent prostate cancer specific survival while preventing
side-effects from radical treatment28
. Especially with the increasing use of PSA as
Introduction and outline
13
a screening tool and therefore a large stage migration to low-risk disease, active
surveillance is appealing both in terms of quality of life as well as cost-effectiveness.
Whether treatment is necessary for all patients therefore remains a vital question.
4. Radiotherapy as primary treatment
When we choose to treat, there is little evidence from randomised controlled trials
that suggests how to treat. As the leading modalities for primary treatment, radical
prostatectomy and radiotherapy do not differ substantially in terms of cancer
control29
. This would make the side-effect profile essential, which might favour
radiotherapy.
There are several developments in the field of radiotherapy that make the treatment
appealing. Using external beam radiotherapy (EBRT), intensity modulated
radiotherapy (IMRT) offers a great advantage over the older 3D conformal
technique (3D-CRT) by increasing the dose delivered to the prostate while sparing
organs at risk (OAR)30,31
. The use of IMRT for treatment of patients with localised
prostate cancer has therefore increased from 0.15% in 2000 to 95.6% in 200832
. A
recent development is that proton therapy is increasingly being adopted, because
of its theoretical advantages in increasing conformality. All therapies show high
biochemical disease free survival (bDFS)29
. However, preliminary results seem
to favour IMRT over proton therapy in terms of treatment-related side-effects,
especially gastrointestinal32,33
.
Furthermore, combining EBRT with brachytherapy, it is possible to boost the
tumour,whichisadvantageousespeciallyforintermediateandhighriskpatients34,35
.
These improvements in accuracy of dose delivery resulted in over 40% of
prostate cancer patients older than 65 year nowadays being treated with EBRT,
brachytherapy or a combination36
.
Where EBRT is traditionally delivered fractionated, brachytherapy can be
delivered in a single procedure. In low and intermediate risk patients, low-dose-
rate (LDR) and high-dose-rate (HDR) brachytherapy can achieve bDFS rates that
are comparable to other primary treatment modalities29,35,37
. Brachytherapy offers a
steep dose decline and high conformality, limiting damage to surrounding organs
at risk and subsequently preventing toxicity. Also the single procedure in which it
can be performed has a great benefit over the many daily fractions with external
beam radiotherapy38
.This makes brachytherapy the most cost-effective primary
treatmentmodalityforprostatecancer39
.Arecentpropensityscoreadjustedanalysis
even indicated that brachytherapy leads to significantly increased bDFS compared
to EBRT for low and intermediate risk patients40
. Unfortunately, brachytherapy as
Introduction and outline
14
a primary treatment modality seems to be declining due to increasing popularity of
other primary (more expensive) treatments, among other factors38,41
.
Another advantage of brachytherapy that was developed in recent years, is a focal
approach to prostate cancer, both with LDR42
and HDR (Netherlands Trial register
number3790andNCT00913939),asopposedtotraditionalwhole-glandtreatment.
This was made possible by increased accuracy of tumour localisation using
multiparametric MRI and (image-guided or systematic) biopsy techniques8,10,16,43,44
.
Also applied with modalities like high intensity focused ultrasound (HIFU)45,46
and
cryotherapy47,48
, the focal approach appears to substantially decrease toxicity rates
while possibly maintaining acceptable cancer control49
. However, longer follow-
up and randomised trials are necessary to ascertain this, because proponents and
opponents considerably differ in their viewpoints49-51
.
Due to this large range of developments in primary radiotherapy and their effects
on prostate cancer management, radiotherapy is increasing as a primary treatment
modality.
5. Recurrent disease after primary radiotherapy
While whole-gland radiotherapy is an established and effective treatment option
for primary tumours, recurrences can still be common. Even in the era of dose
escalation, in which total treatment doses are above 78 Gray (Gy) in the majority
of patients, biochemical relapses occur frequently52-54
. The Kaplan-Meier curves
in figure 2 show the biochemical recurrence rates for the three National Cancer
Comprehensive Network (NCCN) risk groups. Estimated 8-year recurrence rates
of 9.7%, 22.7% and 43.9% for low, intermediate and high risk groups were observed,
respectively. All 2.694 patients were treated with IMRT with a total dose of ≥79.2
Gy, up to maximal 85.6 Gy53
.
The development of biochemical failure is problematic, since it commonly precedes
the development of distant metastases by a median 5.4 years and prostate cancer
specific mortality by a median 10.5 years53
. Even though cancer control rates are
increasing with further dose-escalation, ADT-use and enhanced patient selection,
there will be an increase in absolute numbers of patients with biochemical recurrent
disease because more patients are primarily treated with radiotherapy for prostate
cancer. In addition, the previously discussed rise in PSA-screening will shift the
diagnosis of prostate cancer to an increasingly lower age, creating a group with a
longer life-expectancy after primary treatment and therefore possibly more risk of
developing biochemical failure. Basically, physicians will more often be faced with
biochemical recurrent disease in the future and management of these recurrences
will become an important issue.
Introduction and outline
15
6. The index lesion and the relation with recurrent disease
Biochemical recurrence in prostate cancer often seems to be the result of a localised
process. The data on the exact location of these recurrences is scarce, but in several
series it is suggested that the tumour often regrows at the site of the primary
dominant and usually largest lesion, also called the ‘index lesion’ (figure 3)55-59
.The
evidence is not straightforward, however, with some suggesting tumour regrowth
to occur more multifocal, although organ-confined and even unifocal disease is still
often observed60,61
. Furthermore, salvage radical prostatectomy is not always used
as the reference standard to assess the localised radiorecurrent tumours, thereby
possibly not giving a representable estimate of the exact recurrence location(s).
In addition, patients were often treated with lower doses of radiation than with
current dose-escalation schedules. This could mean that radiorecurrent disease
is currently shifting towards the index lesion because secondary tumour foci are
increasingly successfully treated55,57,58
.
This idea of the index lesion has originated in the primary setting, where it
represents the tumour driving the prognosis of the disease. While prostate cancer
is often a multifocal disease with multiple, often bilateral foci62
, the index lesion is
thought in most cases to make up the largest and most aggressive tumour63-65
.
Figure 2: Biochemical recurrence rates per NCCN risk group53
Low risk 590 547 471 363 263 129 58 19
Int Risk 1289 1129 880 566 354 200 86 24
High Risk 815 653 488 337 226 133 66 29
Low risk
Intermediate Risk
High Risk
PSARecurrenceFreeSurvival
Time (yr)
100%
80%
60%
40%
20%
0%
0 2 4 6 8 10 12 14
Introduction and outline
16
It has even been observed that prostate cancer metastases have a monoclonal
singular origin which could originate in the index lesion, although this is still being
(fiercely) debated66,67
. This idea of the index lesion is tempting, since targeting
therapy to only a focal region of the prostate might reduce toxicity associated
with whole-gland treatment45,46,49,68-71
. As discussed above, treatments in the
primary setting are increasingly targeting partial areas of the gland72,73
, or boosting
dominant intraprostatic lesions74
. Trials are even ongoing in which ‘insignificant’
tumour foci are left untreated45
. Significant developments in staging procedures
using multiparametric-MRI and sophisticated biopsy-techniques make such an
alteration in the way we think of and treat prostate cancer possible. Proponents of
the focal approach compare it to other tumour sites in which this is now common
practice, such as breast and kidney cancer75
. It has even been called a ‘male
lumpectomy’ in the cryotherapy setting48
, equivalent to breast cancer treatment.
For recurrent tumours, this idea of the index lesion and a focal treatment approach
is potentially even more appealing, since secondary tumour foci might have
been successfully treated by the primary radiation. In addition, the decrease in
treatment-related toxicity is potentially much larger in the recurrent treatment
setting. These ideas are gaining increasing international attention and will be
further explored here.
Figure 3: Recurrent prostate tumours. Depicted here as vertical lines (over the horizontal lines indicating
the primary lesion)55
 
Introduction and outline
17
7. Current treatment strategies for recurrent disease
This thesis will focus on the specific subgroup of patients who have locally failed
primary radiotherapy. In prostate cancer, treating localised radiorecurrent prostate
cancer with a curative second approach is referred to as salvage. There are quite
extensive series available in the literature, describing a variety of salvage procedures.
Mostly, these procedures are directed at the entire prostatic volume. The largest
series available to date are those of salvage radical prostatectomy (SRP)76-78
, salvage
cryosurgery (SCS)79-81
, salvage HIFU82,83
and salvage brachytherapy (SBT, both
LDR and HDR)84-87
. All series show decent cancer control rates, with 5-year bDFS
in the range of 50-60% for SRP, SCS and SBT and 40-50% after 3 years for salvage
HIFU88-91
. These techniques therefore have the potential to postpone, or even
prevent the use of ADT, which has been associated with significant cardiovascular,
endocrinologic and sexual toxicity and even mental decline92,93
.
However, a recent overview has shown that salvage therapies are not frequently
adopted, with approximately 98% of patients receiving ADT as subsequent
treatment for recurrent disease. Only 2% of patients were found to be treated with
a form of curatively intended salvage94
. This was also observed in an analysis of
the large (UCSF) Cancer of the Prostate Strategic Urological Research Endeavor
(CaPSURE) database95
.
A reason to refrain from salvage therapies is that they are mostly performed
on a whole-gland basis88,90,91,96
. Because of prior difficulties in the assessment of
recurrences, whole gland approaches were (and are still often) the only way to make
sure residual disease is fully targeted. The downside to a whole-gland approach is
the severe toxicity associated with these procedures. After primary radiotherapy,
the prostate becomes fibrotic and OAR surrounding the prostate such as the
rectal wall, bladder neck and urethra, show similar changes85,97
. These fibrotic
changes impair the normal repair capacities of these structures, making salvage
surgical approaches difficult and cumulative radiation from SBT hazardous. High
percentages of toxicity have been found with all salvage modalities, although with
a wide spread across series. Summarised, up to a third of patients can experience
a combination of severe (≥grade 3) genitourinary (GU) and gastro-intestinal (GI)
toxicity, requiring surgical intervention to resolve89,91,96,98
. Erectile dysfunction is
poorly reported and frequently pre-existent, but often found to be almost universal
after salvage procedures88,96,98
. These severe side-effects are likely to contribute
to the previously mentioned 2% of patients who are treated with some form of
salvage. Salvage has for these reasons remained a treatment generally offered only
in specialised, high volume centres.
Introduction and outline
18
8. Focal salvage
Due to the severe toxicity rates associated with whole-gland salvage in combination
with the developments in accurate staging and the attention on focally targeting
prostate cancer, the hypothesis of a focal approach in the salvage setting is being
developed. With the evidence that recurrences are often localised at the primary
index lesion and secondary tumour foci are successfully treated by the primary
radiation, the rationale for a focal approach might be even more potent in the
salvage than in the primary setting. A focal approach could theoretically spare
a substantial part of the prostate and with this, prevent damage to surrounding
OAR. However, this procedure is technically challenging and the work-up to assess
focal recurrences is intensive and requires a high level of specialised assessment99
.
Therefore, focal salvage series are limited in the literature and performed only
in specialised centres. A variety of techniques are now described in some pilot
series, most importantly cryotherapy (n=10-91)100-103
, HIFU (n=39-48)104,105
and Iodine-125 (LDR) brachytherapy (n=15-25)106-108
. Most of these series
adopt a multimodality approach in assessing recurrent disease. This consists of
multiparametric MRI: T2-weighted, dynamic contrast enhanced (DCE), diffusion
weighted imaging (DWI) and sometimes spectroscopy (MRSI). Furthermore,
tumour location is confirmed by either systematic transrectal ultrasound guided
biopsies, or template prostate mapping biopsies (TTPM) in one series104
. The
extent of ablation varies from completely focal with a specified margin to hemi or
zonal, which is also referred to as a ‘dog leg’ or ‘hockey stick’ ablation (Figure 4)73
.
Results from focal salvage series are preliminary, but seem favourable. Reported
focal salvage 1, 2, 3 and 5- year bDFS rates are 69%-100%, 49%-100%, 50%-72.4%
and 46.5%-54.4%, respectively101-108
. These are in line with bDFS-outcomes from
 
Targeted ablation Hemiablation Zonal ablation
Figure 4: Different focal ablation methods for the primary and salvage setting73
.
Introduction and outline
19
the whole-gland setting. However, a selection bias towards a more favourable focal
salvage patient group cannot be excluded based on these observational studies.
Randomised trials, or (less favourably) propensity-score adjusted observational
cohorts will have to assess whether there are differences in cancer control between
a whole-gland and focal salvage approach.
Nonetheless, even if cancer control rates are in favour of whole-gland salvage, the
decrease in toxicity with focal salvage could still be a reason to refrain from a whole-
gland approach. Toxicity results from focal salvage are favourable, with most studies
reporting side-effects which are self-limiting or treatable with medication (grade
1 and 2)101,103,106,107
. Some series report higher toxicity rates, which sometimes need
surgical intervention to resolve104,108
. The extent of treatment could have influenced
these results, with one study targeting the entire peripheral zone108
. If toxicity results
are favourable compared to whole-gland approaches, repeating focal salvage could
even be an option if patients initially fail retreatment. This has been successfully
done in one of the focal SBT series106
and focal salvage cryotherapy series100
.
9. Problems with current whole-gland and focal salvage I-125
brachytherapy
Brachytherapy is often adopted as a salvage modality, both in the whole-gland and
focal savage setting. To perform SBT in a safe and effective way, dose constraints
and selection criteria related to cancer control are of vital importance. However,
these are not available in the current literature.
Dose constraints are usually adopted from the primary setting. However, these
restrictions might be too lenient because of previous radiation damage to
surrounding organs at risk109-113
. It is vital that there are constraints found for SBT
below which toxicity is reduced to a minimum, in a way equivalent to the research
which is available for primary brachytherapy111,112,114
.
Selection criteria in relation to cancer control are available from multivariable
analyses performed in larger whole-gland salvage cohorts using SRP, SCS and
HIFU76,79,83
. For whole-gland SBT, these are less well defined, due to smaller series
with limited events to which prognostic determinants can be related85-87,115
.
Also, predictive factors are often categorised (leading to information loss), missing
data is not described and no prediction models are available to obtain adequate
individual prognoses. These problems are also observed in the other salvage
modalities. Furthermore, due to the limited focal salvage data so far, there has not
been any uni- or multivariable modelling done to identify factors related to cancer
control. For these reasons, selection of patients for whole-gland and especially focal
Introduction and outline
20
salvage is now often performed on the basis of determinants related to prognosis in
the primary setting or on expert opinion.
Conclusively, to this day there are no reliable dose constraints, selection criteria
and prediction models developed specifically for SBT. They are frequently adopted
from primary BT data and do not take into account issues such as previous
radiation damage to OAR or the clinical profile of patients.
10. Outline
This thesis will focus on cancer control and toxicity regarding whole-gland salvage
modalities and focal salvage Iodine-125 brachytherapy and HIFU.
Chapter 1 demonstrates that whole-gland salvage modalities adopted in the
Netherlands nowadays are associated with high failure rates, probably because of
insufficient assessment of localised disease and exclusion of metastases.
Also, significant toxicity rates are observed, which is probably due to targeting the
entire prostatic volume. To prevent the high toxicity rates of these whole-gland
approaches while maintaining cancer control, focal salvage might be a viable
solution. The literature provides us with small series assessing different focal
salvage modalities. Chapter 2 summarises literature results of focal and whole-
gland salvage and compares cancer control and toxicity.
A focal salvage approach using I-125 (LDR) brachytherapy has further been
evaluated at our department. Twenty patients with a biochemical recurrence after
primary radiotherapy (EBRT or I-125 brachytherapy) were treated. Cancer control,
toxicity and quality of life of this cohort are evaluated in chapter 3.
The whole-gland I-125 brachytherapy cohort from chapter 1 and the focal salvage
cohort from chapter 3 were further evaluated regarding dosimetry and toxicity
of both the genitourinary (GU) and gastro-intestinal (GI) system. With these
dosimetry differences it was possible to construct dose constraints to prevent late
severe GU and GI toxicity (chapters 4 and 5, respectively). Dose constraints for
salvage I-125 brachytherapy are important for future adoption of this technique,
since constraints for the primary setting might be too lenient in salvage patients
because of previous radiation damage to surrounding organs at risk.
Chapters 6, 7 and 8 are all related to prediction models to improve selection of
patients for both whole-gland and focal salvage. Selection is often performed based
on characteristics associated with failure in the primary setting. However, whether
these characteristics have the same predictive value in the salvage setting is mostly
unknown. No prediction models to quantify the exact effects of pre-treatment
characteristics are available for salvage. For the larger whole-gland salvage series,
Introduction and outline
21
there are some multivariable models assessing the effects of certain characteristics.
For SBT, these models are based on small datasets with often limited events,
categorisation of predictors, insufficient description of missing data and other
problems which lead to an inaccurate assessment of the effect of prognostic factors.
To overcome these problems, chapters 6 and 7 use the most recent methodological
guidelines to quantify the effect of prognostic factors on both biochemical failure
(chapter 6) and mortality (chapter 7, both prostate cancer specific and overall
mortality). With these prediction models, a more accurate selection of whole-
gland SBT patients is possible.
Nevertheless, these predictors are assessed for whole-gland SBT and might have
a different relation with the outcome in focal salvage. The focal SBT cohort from
chapter 3 was too limited to create a prediction model. Therefore, in cooperation
with the University College London Hospitals (UCLH) Department of Urology,
a prediction model was created based on their focal salvage HIFU cohort after
EBRT-failure. This prediction model was based on a substantially larger cohort and
therefore the creation of a clinically useful risk score was possible. These results are
described in chapter 8.
Lastly, if focal salvage leads to equivalent cancer control rates and less toxicity
than whole-gland salvage, cost-effectiveness is an important aspect which needs to
be assessed. Chapter 9 compares the results of the whole-gland and focal salvage
I-125 brachytherapy cohorts in terms of cost-effectiveness. The limitations of the
described research and future perspectives with regard to salvage are discussed
after these chapters in the general discussion.
Introduction and outline
22
References
1.	 Siegel RL, Sahar L, Portier KM, Ward EM, Jemal A. Cancer death rates in US congressional
districts. CA Cancer J Clin. 2015. doi: 10.3322/caac.21292 [doi].
2.	 Integraal kankercentrum Nederland. Incidentie prostaatkanker. http://www.cijfersoverkanker.
nl/selecties/Incidentie_prostaatkanker/img54d0ecdf1c883. Updated 2014. Accessed 09/25, 2015.
3.	 Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: Sources,
methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359-86. doi:
10.1002/ijc.29210 [doi].
4.	 Integraal Kankercentrum Nederland. Richtlijn prostaatcarcinoom. http://www.oncoline.nl/
richtlijn/doc/index.php?type=pda&richtlijn_id=934. Updated 2014. Accessed 09/01, 2015.
5.	 Partin AW, Mangold LA, Lamm DM, Walsh PC, Epstein JI, Pearson JD. Contemporary update
of prostate cancer staging nomograms (partin tables) for the new millennium. Urology.
2001;58(6):843-848. doi: S0090429501014418 [pii].
6.	 Barentsz JO, Richenberg J, Clements R, et al. ESUR prostate MR guidelines 2012. Eur Radiol.
2012;22(4):746-757. doi: 10.1007/s00330-011-2377-y [doi].
7.	 Umbehr MH, Muntener M, Hany T, Sulser T, Bachmann LM. The role of 11C-choline and
18F-fluorocholine positron emission tomography (PET) and PET/CT in prostate cancer:
A systematic review and meta-analysis. Eur Urol. 2013;64(1):106-117. doi: 10.1016/j.
eururo.2013.04.019 [doi].
8.	 Futterer JJ, Briganti A, De Visschere P, et al. Can clinically significant prostate cancer be detected
with multiparametric magnetic resonance imaging? A systematic review of the literature. Eur
Urol. 2015. doi: S0302-2838(15)00036-6 [pii].
9.	 Hamoen EH, de Rooij M, Witjes JA, Barentsz JO, Rovers MM. Use of the prostate imaging
reporting and data system (PI-RADS) for prostate cancer detection with multiparametric
magnetic resonance imaging: A diagnostic meta-analysis. Eur Urol. 2015;67(6):1112-1121. doi:
10.1016/j.eururo.2014.10.033 [doi].
10.	 de Rooij M, Hamoen EH, Witjes JA, Barentsz JO, Rovers MM. Accuracy of magnetic resonance
imaging for local staging of prostate cancer: A diagnostic meta-analysis. Eur Urol. 2015. doi:
S0302-2838(15)00674-0 [pii].
11.	 Onur R, Littrup PJ, Pontes JE, Bianco FJ,Jr. Contemporary impact of transrectal ultrasound lesions
for prostate cancer detection. J Urol. 2004;172(2):512-514. doi: S0022-5347(05)61673-4 [pii].
12.	 Schroder FH, van der Maas P, Beemsterboer P, et al. Evaluation of the digital rectal examination
as a screening test for prostate cancer. rotterdam section of the european randomized study of
screening for prostate cancer. J Natl Cancer Inst. 1998;90(23):1817-1823.
13.	 Abuzallouf S, Dayes I, Lukka H. Baseline staging of newly diagnosed prostate cancer: A summary
of the literature. J Urol. 2004;171(6 Pt 1):2122-2127. doi: 00005392-200406000-00006 [pii].
14.	 Evangelista L, Zattoni F, Guttilla A, et al. Choline PET or PET/CT and biochemical relapse of
prostate cancer: A systematic review and meta-analysis. Clin Nucl Med. 2013;38(5):305-314. doi:
10.1097/RLU.0b013e3182867f3c [doi].
Introduction and outline
23
15.	 Pokorny MR, de Rooij M, Duncan E, et al. Prospective study of diagnostic accuracy comparing
prostate cancer detection by transrectal ultrasound-guided biopsy versus magnetic resonance
(MR) imaging with subsequent MR-guided biopsy in men without previous prostate biopsies. Eur
Urol. 2014;66(1):22-29. doi: 10.1016/j.eururo.2014.03.002 [doi].
16.	 Siddiqui MM, Rais-Bahrami S, Turkbey B, et al. Comparison of MR/ultrasound fusion-guided
biopsywithultrasound-guidedbiopsyforthediagnosisofprostatecancer.JAMA.2015;313(4):390-
397. doi: 10.1001/jama.2014.17942 [doi].
17.	 Valerio M, Anele C, Charman SC, et al. Transperineal template prostate mapping biopsies: An
evaluation of different protocols in detection of clinically significant prostate cancer. BJU Int.
2015. doi: 10.1111/bju.13306 [doi].
18.	 Moore CM, Robertson NL, Arsanious N, et al. Image-guided prostate biopsy using magnetic
resonance imaging-derived targets: A systematic review. Eur Urol. 2013;63(1):125-140. doi:
10.1016/j.eururo.2012.06.004 [doi].
19.	 Wilt TJ, Brawer MK, Jones KM, et al. Radical prostatectomy versus observation for localised
prostate cancer. N Engl J Med. 2012;367(3):203-213. doi: 10.1056/NEJMoa1113162 [doi].
20.	 Bill-Axelson A, Holmberg L, Garmo H, et al. Radical prostatectomy or watchful waiting in early
prostate cancer. N Engl J Med. 2014;370(10):932-942. doi: 10.1056/NEJMoa1311593 [doi].
21.	 Schroder FH, Hugosson J, Roobol MJ, et al. Screening and prostate-cancer mortality in a
randomizedeuropeanstudy.NEnglJMed.2009;360(13):1320-1328.doi:10.1056/NEJMoa0810084
[doi].
22.	 Schroder FH, Hugosson J, Roobol MJ, et al. Prostate-cancer mortality at 11 years of follow-up. N
Engl J Med. 2012;366(11):981-990. doi: 10.1056/NEJMoa1113135 [doi].
23.	 Schroder FH, Hugosson J, Roobol MJ, et al. Screening and prostate cancer mortality: Results of
the european randomised study of screening for prostate cancer (ERSPC) at 13 years of follow-up.
Lancet. 2014;384(9959):2027-2035. doi: 10.1016/S0140-6736(14)60525-0 [doi].
24.	 Andriole GL, Crawford ED, Grubb RL,3rd, et al. Prostate cancer screening in the randomized
prostate, lung, colorectal, and ovarian cancer screening trial: Mortality results after 13 years of
follow-up. J Natl Cancer Inst. 2012;104(2):125-132. doi: 10.1093/jnci/djr500 [doi].
25.	 Dahm P, Neuberger M, Ilic D. Screening for prostate cancer: Shaping the debate on benefits and
harms. Cochrane Database Syst Rev. 2013;9:ED000067. doi: org/10.1002/14651858.ED000067
[doi].
26.	 Ilic D, Neuberger MM, Djulbegovic M, Dahm P. Screening for prostate cancer. Cochrane Database
Syst Rev. 2013;1:CD004720. doi: 10.1002/14651858.CD004720.pub3 [doi].
27.	 Resnick MJ, Koyama T, Fan KH, et al. Long-term functional outcomes after treatment for localised
prostate cancer. N Engl J Med. 2013;368(5):436-445. doi: 10.1056/NEJMoa1209978 [doi].
28.	 Klotz L, Zhang L, Lam A, Nam R, Mamedov A, Loblaw A. Clinical results of long-term follow-up
of a large, active surveillance cohort with localised prostate cancer. J Clin Oncol. 2010;28(1):126-
131. doi: 10.1200/JCO.2009.24.2180 [doi].
Introduction and outline
24
29.	 Grimm P, Billiet I, Bostwick D, et al. Comparative analysis of prostate-specific antigen free survival
outcomes for patients with low, intermediate and high risk prostate cancer treatment by radical
therapy. results from the prostate cancer results study group. BJU Int. 2012;109 Suppl 1:22-29. doi:
10.1111/j.1464-410X.2011.10827.x [doi].
30.	 Cahlon O, Hunt M, Zelefsky MJ. Intensity-modulated radiation therapy: Supportive data for
prostate cancer. Semin Radiat Oncol. 2008;18(1):48-57. doi: S1053-4296(07)00079-3 [pii].
31.	 Guckenberger M, Flentje M. Intensity-modulated radiotherapy (IMRT) of localised prostate
cancer: A review and future perspectives. Strahlenther Onkol. 2007;183(2):57-62. doi: 10.1007/
s00066-007-1631-3 [doi].
32.	 Sheets NC, Goldin GH, Meyer AM, et al. Intensity-modulated radiation therapy, proton therapy,
or conformal radiation therapy and morbidity and disease control in localised prostate cancer.
JAMA. 2012;307(15):1611-1620. doi: 10.1001/jama.2012.460 [doi].
33.	 Kim S, Shen S, Moore DF, et al. Late gastrointestinal toxicities following radiation therapy for
prostate cancer. Eur Urol. 2011;60(5):908-916. doi: 10.1016/j.eururo.2011.05.052 [doi].
34.	 Hoskin PJ, Rojas AM, Bownes PJ, Lowe GJ, Ostler PJ, Bryant L. Randomised trial of external beam
radiotherapy alone or combined with high-dose-rate brachytherapy boost for localised prostate
cancer. Radiother Oncol. 2012;103(2):217-222. doi: 10.1016/j.radonc.2012.01.007 [doi].
35.	 Morton GC, Hoskin PJ. Brachytherapy: Current status and future strategies -- can high dose rate
replace low dose rate and external beam radiotherapy? Clin Oncol (R Coll Radiol). 2013;25(8):474-
482. doi: 10.1016/j.clon.2013.04.009 [doi].
36.	 DeSantis CE, Lin CC, Mariotto AB, et al. Cancer treatment and survivorship statistics, 2014. CA
Cancer J Clin. 2014;64(4):252-271. doi: 10.3322/caac.21235 [doi].
37.	 Morris WJ, Spadinger I, Keyes M, Hamm J, McKenzie M, Pickles T. Whole prostate D90 and
V100: A dose-response analysis of 2000 consecutive (125)I monotherapy patients. Brachytherapy.
2014;13(1):32-41. doi: 10.1016/j.brachy.2013.08.006 [doi].
38.	 Petereit DG, Frank SJ, Viswanathan AN, et al. Brachytherapy: Where has it gone? J Clin Oncol.
2015;33(9):980-982. doi: 10.1200/JCO.2014.59.8128 [doi].
39.	 Hayes JH, Ollendorf DA, Pearson SD, et al. Observation versus initial treatment for men
with localised, low-risk prostate cancer: A cost-effectiveness analysis. Ann Intern Med.
2013;158(12):853-860. doi: 10.7326/0003-4819-158-12-201306180-00002 [doi].
40.	 Smith GD, Pickles T, Crook J, et al. Brachytherapy improves biochemical failure-free survival in
low- and intermediate-risk prostate cancer compared with conventionally fractionated external
beam radiation therapy: A propensity score matched analysis. Int J Radiat Oncol Biol Phys.
2015;91(3):505-516. doi: 10.1016/j.ijrobp.2014.11.018 [doi].
41.	 Martin JM, Handorf EA, Kutikov A, et al. The rise and fall of prostate brachytherapy: Use of
brachytherapy for the treatment of localised prostate cancer in the national cancer data base.
Cancer. 2014;120(14):2114-2121. doi: 10.1002/cncr.28697 [doi].
42.	 Nguyen PL, Chen MH, Zhang Y, et al. Updated results of magnetic resonance imaging guided
partial prostate brachytherapy for favorable risk prostate cancer: Implications for focal therapy. J
Urol. 2012;188(4):1151-1156. doi: 10.1016/j.juro.2012.06.010 [doi].
Introduction and outline
25
43.	 Groenendaal G, Moman MR, Korporaal JG, et al. Validation of functional imaging with pathology
for tumour delineation in the prostate. Radiother Oncol. 2010;94(2):145-150. doi: 10.1016/j.
radonc.2009.12.034 [doi].
44.	 Al-Qaisieh B, Mason J, Bownes P, et al. Dosimetry modeling for focal low-dose-rate prostate
brachytherapy. Int J Radiat Oncol Biol Phys. 2015;92(4):787-793. doi: 10.1016/j.ijrobp.2015.02.043
[doi].
45.	 Ahmed HU, Dickinson L, Charman S, et al. Focal ablation targeted to the index lesion in
multifocal localised prostate cancer: A prospective development study. Eur Urol. 2015. doi: S0302-
2838(15)00073-1 [pii].
46.	 Ahmed HU, Hindley RG, Dickinson L, et al. Focal therapy for localised unifocal and multifocal
prostate cancer: A prospective development study. Lancet Oncol. 2012;13(6):622-632. doi:
10.1016/S1470-2045(12)70121-3 [doi].
47.	 Bahn D, de Castro Abreu AL, Gill IS, et al. Focal cryotherapy for clinically unilateral, low-
intermediate risk prostate cancer in 73 men with a median follow-up of 3.7 years. Eur Urol.
2012;62(1):55-63. doi: 10.1016/j.eururo.2012.03.006 [doi].
48.	 Onik G, Vaughan D, Lotenfoe R, Dineen M, Brady J. "Male lumpectomy": Focal therapy for prostate
cancer using cryoablation. Urology. 2007;70(6 Suppl):16-21. doi: 10.1016/j.urology.2007.06.001
[doi].
49.	 Valerio M, Ahmed HU, Emberton M, et al. The role of focal therapy in the management of
localised prostate cancer: A systematic review. Eur Urol. 2014;66(4):732-751. doi: 10.1016/j.
eururo.2013.05.048 [doi].
50.	 Giannarini G, Gandaglia G, Montorsi F, Briganti A. Will focal therapy remain only an attractive
illusion for the primary treatment of prostate cancer? J Clin Oncol. 2014;32(13):1299-1301. doi:
10.1200/JCO.2013.54.8214 [doi].
51.	 Valerio M, Emberton M, Ahmed HU. Focal therapy will become a standard option for
selected men with localised prostate cancer. J Clin Oncol. 2014;32(32):3680-3681. doi: 10.1200/
JCO.2014.56.7792 [doi].
52.	 Cahlon O, Zelefsky MJ, Shippy A, et al. Ultra-high dose (86.4 gy) IMRT for localised prostate
cancer: Toxicity and biochemical outcomes. Int J Radiat Oncol Biol Phys. 2008;71(2):330-337. doi:
10.1016/j.ijrobp.2007.10.004 [doi].
53.	 ZumstegZS,SprattDE,RomesserPB,etal.Thenaturalhistoryandpredictorsofoutcomefollowing
biochemical relapse in the dose escalation era for prostate cancer patients undergoing definitive
external beam radiotherapy. Eur Urol. 2015;67(6):1009-1016. doi: 10.1016/j.eururo.2014.09.028
[doi].
54.	 Zelefsky MJ, Pei X, Chou JF, et al. Dose escalation for prostate cancer radiotherapy: Predictors of
long-term biochemical tumour control and distant metastases-free survival outcomes. Eur Urol.
2011;60(6):1133-1139. doi: 10.1016/j.eururo.2011.08.029 [doi].
55.	 Cellini N, Morganti AG, Mattiucci GC, et al. Analysis of intraprostatic failures in patients treated
with hormonal therapy and radiotherapy: Implications for conformal therapy planning. Int J
Radiat Oncol Biol Phys. 2002;53(3):595-599. doi: S0360301602027955 [pii].
Introduction and outline
26
56.	 Pucar D, Hricak H, Shukla-Dave A, et al. Clinically significant prostate cancer local recurrence
after radiation therapy occurs at the site of primary tumour: Magnetic resonance imaging and
step-section pathology evidence. Int J Radiat Oncol Biol Phys. 2007;69(1):62-69. doi: S0360-
3016(07)00674-8 [pii].
57.	 Arrayeh E, Westphalen AC, Kurhanewicz J, et al. Does local recurrence of prostate cancer after
radiation therapy occur at the site of primary tumour? results of a longitudinal MRI and MRSI
study. Int J Radiat Oncol Biol Phys. 2012;82(5):e787-93. doi: 10.1016/j.ijrobp.2011.11.030 [doi].
58.	 Menard C, Iupati D, Publicover J, et al. MR-guided prostate biopsy for planning of focal salvage
after radiation therapy. Radiology. 2015;274(1):181-191. doi: 10.1148/radiol.14122681 [doi].
59.	 Cheng L, Cheville JC, Pisansky TM, et al. Prevalence and distribution of prostatic intraepithelial
neoplasia in salvage radical prostatectomy specimens after radiation therapy. Am J Surg Pathol.
1999;23(7):803-808.
60.	 Huang WC, Kuroiwa K, Serio AM, et al. The anatomical and pathological characteristics of
irradiated prostate cancers may influence the oncological efficacy of salvage ablative therapies. J
Urol. 2007;177(4):1324-9; quiz 1591. doi: S0022-5347(06)03110-7 [pii].
61.	 Leibovici D, Chiong E, Pisters LL, et al. Pathological characteristics of prostate cancer recurrence
after radiation therapy: Implications for focal salvage therapy. J Urol. 2012;188(1):98-102. doi:
10.1016/j.juro.2012.02.2571 [doi].
62.	 Andreoiu M, Cheng L. Multifocal prostate cancer: Biologic, prognostic, and therapeutic
implications. Hum Pathol. 2010;41(6):781-793. doi: 10.1016/j.humpath.2010.02.011 [doi].
63.	 Ahmed HU. The index lesion and the origin of prostate cancer. N Engl J Med. 2009;361(17):1704-
1706. doi: 10.1056/NEJMcibr0905562 [doi].
64.	 Stamey TA, McNeal JM, Wise AM, Clayton JL. Secondary cancers in the prostate do not determine
PSA biochemical failure in untreated men undergoing radical retropubic prostatectomy. Eur Urol.
2001;39 Suppl 4:22-23. doi: 52577 [pii].
65.	 Wise AM, Stamey TA, McNeal JE, Clayton JL. Morphologic and clinical significance of
multifocal prostate cancers in radical prostatectomy specimens. Urology. 2002;60(2):264-269. doi:
S0090429502017284 [pii].
66.	 Liu W, Laitinen S, Khan S, et al. Copy number analysis indicates monoclonal origin of lethal
metastatic prostate cancer. Nat Med. 2009;15(5):559-565. doi: 10.1038/nm.1944 [doi].
67.	 Haffner MC, Mosbruger T, Esopi DM, et al. Tracking the clonal origin of lethal prostate cancer. J
Clin Invest. 2013;123(11):4918-4922. doi: 10.1172/JCI70354 [doi].
68.	 Ahmed HU, Pendse D, Illing R, Allen C, van der Meulen JH, Emberton M. Will focal therapy
become a standard of care for men with localised prostate cancer? Nat Clin Pract Oncol.
2007;4(11):632-642. doi: ncponc0959 [pii].
69.	 Eggener SE, Scardino PT, Carroll PR, et al. Focal therapy for localised prostate cancer: A critical
appraisal of rationale and modalities. J Urol. 2007;178(6):2260-2267. doi: S0022-5347(07)02162-3
[pii].
Introduction and outline
27
70.	 Kasivisvanathan V, Emberton M, Ahmed HU. Focal therapy for prostate cancer: Rationale
and treatment opportunities. Clin Oncol (R Coll Radiol). 2013;25(8):461-473. doi: 10.1016/j.
clon.2013.05.002 [doi].
71.	 Klotz L, Emberton M. Management of low risk prostate cancer-active surveillance and focal
therapy. Nat Rev Clin Oncol. 2014;11(6):324-334. doi: 10.1038/nrclinonc.2014.73 [doi].
72.	 Donaldson IA, Alonzi R, Barratt D, et al. Focal therapy: Patients, interventions, and outcomes--a
report from a consensus meeting. Eur Urol. 2015;67(4):771-777. doi: 10.1016/j.eururo.2014.09.018
[doi].
73.	 van den Bos W, Muller BG, Ahmed H, et al. Focal therapy in prostate cancer: International
multidisciplinary consensus on trial design. Eur Urol. 2014;65(6):1078-1083. doi: 10.1016/j.
eururo.2014.01.001 [doi].
74.	 Lips IM, van der Heide UA, Haustermans K, et al. Single blind randomized phase III trial to
investigate the benefit of a focal lesion ablative microboost in prostate cancer (FLAME-trial): Study
protocol for a randomized controlled trial. Trials. 2011;12:255-6215-12-255. doi: 10.1186/1745-
6215-12-255 [doi].
75.	 Ahmed HU. Introduction--targeting the lesion, not the organ. Urol Oncol. 2014;32(6):901-902.
doi: 10.1016/j.urolonc.2014.06.011 [doi].
76.	 Chade DC, Shariat SF, Cronin AM, et al. Salvage radical prostatectomy for radiation-recurrent
prostate cancer: A multi-institutional collaboration. Eur Urol. 2011;60(2):205-210. doi: 10.1016/j.
eururo.2011.03.011 [doi].
77.	 Chade DC, Eastham J, Graefen M, et al. Cancer control and functional outcomes of salvage radical
prostatectomy for radiation-recurrent prostate cancer: A systematic review of the literature. Eur
Urol. 2012;61(5):961-971. doi: 10.1016/j.eururo.2012.01.022 [doi].
78.	 Paparel P, Cronin AM, Savage C, Scardino PT, Eastham JA. Oncologic outcome and patterns
of recurrence after salvage radical prostatectomy. Eur Urol. 2009;55(2):404-410. doi: 10.1016/j.
eururo.2008.07.007 [doi].
79.	 Wenske S, Quarrier S, Katz AE. Salvage cryosurgery of the prostate for failure after primary
radiotherapy or cryosurgery: Long-term clinical, functional, and oncologic outcomes in a large
cohort at a tertiary referral centre. Eur Urol. 2013;64(1):1-7. doi: 10.1016/j.eururo.2012.07.008
[doi].
80.	 Williams AK, Martinez CH, Lu C, Ng CK, Pautler SE, Chin JL. Disease-free survival following
salvage cryotherapy for biopsy-proven radio-recurrent prostate cancer. Eur Urol. 2011;60(3):405-
410. doi: 10.1016/j.eururo.2010.12.012 [doi].
81.	 Spiess PE, Katz AE, Chin JL, et al. A pretreatment nomogram predicting biochemical failure
after salvage cryotherapy for locally recurrent prostate cancer. BJU Int. 2010;106(2):194-198. doi:
10.1111/j.1464-410X.2009.09078.x [doi].
82.	 Crouzet S, Murat FJ, Pommier P, et al. Locally recurrent prostate cancer after initial radiation
therapy: Early salvage high-intensity focused ultrasound improves oncologic outcomes. Radiother
Oncol. 2012;105(2):198-202. doi: 10.1016/j.radonc.2012.09.014 [doi].
Introduction and outline
28
83.	 Murat FJ, Poissonnier L, Rabilloud M, et al. Mid-term results demonstrate salvage high-
intensity focused ultrasound (HIFU) as an effective and acceptably morbid salvage treatment
option for locally radiorecurrent prostate cancer. Eur Urol. 2009;55(3):640-647. doi: 10.1016/j.
eururo.2008.04.091 [doi].
84.	 Chen CP, Weinberg V, Shinohara K, et al. Salvage HDR brachytherapy for recurrent prostate
cancer after previous definitive radiation therapy: 5-year outcomes. Int J Radiat Oncol Biol Phys.
2013;86(2):324-329. doi: 10.1016/j.ijrobp.2013.01.027 [doi].
85.	 Grado GL, Collins JM, Kriegshauser JS, et al. Salvage brachytherapy for localised prostate cancer
after radiotherapy failure. Urology. 1999;53(1):2-10. doi: S0090-4295(98)00492-0 [pii].
86.	 Moman MR, van der Poel HG, Battermann JJ, Moerland MA, van Vulpen M. Treatment outcome
and toxicity after salvage 125-I implantation for prostate cancer recurrences after primary 125-I
implantation and external beam radiotherapy. Brachytherapy. 2010;9(2):119-125. doi: 10.1016/j.
brachy.2009.06.007 [doi].
87.	 Burri RJ, Stone NN, Unger P, Stock RG. Long-term outcome and toxicity of salvage brachytherapy
for local failure after initial radiotherapy for prostate cancer. Int J Radiat Oncol Biol Phys.
2010;77(5):1338-1344. doi: 10.1016/j.ijrobp.2009.06.061 [doi].
88.	 Punnen S, Cooperberg MR, D'Amico AV, et al. Management of biochemical recurrence
after primary treatment of prostate cancer: A systematic review of the literature. Eur Urol.
2013;64(6):905-915. doi: 10.1016/j.eururo.2013.05.025 [doi].
89.	 Parekh A, Graham PL, Nguyen PL. Cancer control and complications of salvage local therapy
after failure of radiotherapy for prostate cancer: A systematic review. Semin Radiat Oncol.
2013;23(3):222-234. doi: 10.1016/j.semradonc.2013.01.006 [doi].
90.	 Kimura M, Mouraviev V, Tsivian M, Mayes JM, Satoh T, Polascik TJ. Current salvage methods for
recurrent prostate cancer after failure of primary radiotherapy. BJU Int. 2010;105(2):191-201. doi:
10.1111/j.1464-410X.2009.08715.x [doi].
91.	 Nguyen PL, D'Amico AV, Lee AK, Suh WW. Patient selection, cancer control, and complications
after salvage local therapy for postradiation prostate-specific antigen failure: A systematic review
of the literature. Cancer. 2007;110(7):1417-1428. doi: 10.1002/cncr.22941 [doi].
92.	 Heidenreich A, Bastian PJ, Bellmunt J, et al. EAU guidelines on prostate cancer. part II: Treatment
of advanced, relapsing, and castration-resistant prostate cancer. Eur Urol. 2014;65(2):467-479. doi:
10.1016/j.eururo.2013.11.002 [doi].
93.	 Nguyen PL, Alibhai SM, Basaria S, et al. Adverse effects of androgen deprivation therapy and
strategies to mitigate them. Eur Urol. 2015;67(5):825-836. doi: 10.1016/j.eururo.2014.07.010 [doi].
94.	 Tran H, Kwok J, Pickles T, Tyldesley S, Black PC. Underutilization of local salvage therapy
after radiation therapy for prostate cancer. Urol Oncol. 2014;32(5):701-706. doi: 10.1016/j.
urolonc.2013.12.014 [doi].
95.	 Agarwal PK, Sadetsky N, Konety BR, Resnick MI, Carroll PR, Cancer of the Prostate Strategic
Urological Research Endeavor (CaPSURE). Treatment failure after primary and salvage therapy
for prostate cancer: Likelihood, patterns of care, and outcomes. Cancer. 2008;112(2):307-314. doi:
10.1002/cncr.23161 [doi].
Introduction and outline
29
96.	 Alongi F, De Bari B, Campostrini F, et al. Salvage therapy of intraprostatic failure after radical
external-beamradiotherapyforprostatecancer:Areview.CritRevOncolHematol.2013;88(3):550-
563. doi: 10.1016/j.critrevonc.2013.07.009 [doi].
97.	 Marks LB, Carroll PR, Dugan TC, Anscher MS. The response of the urinary bladder, urethra, and
ureter to radiation and chemotherapy. Int J Radiat Oncol Biol Phys. 1995;31(5):1257-1280. doi:
0360-3016(94)00431-J [pii].
98.	 Peters M, Moman MR, van der Poel HG, et al. Patterns of outcome and toxicity after salvage
prostatectomy, salvage cryosurgery and salvage brachytherapy for prostate cancer recurrences after
radiation therapy: A multi-center experience and literature review. World J Urol. 2013;31(2):403-
409. doi: 10.1007/s00345-012-0928-8 [doi].
99.	 Crehange G, Roach M,3rd, Martin E, et al. Salvage reirradiation for locoregional failure after
radiation therapy for prostate cancer: Who, when, where and how? Cancer Radiother. 2014;18(5-
6):524-534. doi: 10.1016/j.canrad.2014.07.153 [doi].
100.	 Bomers JG, Yakar D, Overduin CG, et al. MR imaging-guided focal cryoablation in patients with
recurrent prostate cancer. Radiology. 2013;268(2):451-460. doi: 10.1148/radiol.13121291 [doi].
101.	 de Castro Abreu AL, Bahn D, Leslie S, et al. Salvage focal and salvage total cryoablation for locally
recurrent prostate cancer after primary radiation therapy. BJU Int. 2013;112(3):298-307. doi:
10.1111/bju.12151 [doi].
102.	 Eisenberg ML, Shinohara K. Partial salvage cryoablation of the prostate for recurrent
prostate cancer after radiotherapy failure. Urology. 2008;72(6):1315-1318. doi: 10.1016/j.
urology.2008.03.040 [doi].
103.	 Li YH, Elshafei A, Agarwal G, Ruckle H, Powsang J, Jones JS. Salvage focal prostate cryoablation
for locally recurrent prostate cancer after radiotherapy: Initial results from the cryo on-line data
registry. Prostate. 2015;75(1):1-7. doi: 10.1002/pros.22881 [doi].
104.	 Ahmed HU, Cathcart P, McCartan N, et al. Focal salvage therapy for localised prostate cancer
recurrence after external beam radiotherapy: A pilot study. Cancer. 2012;118(17):4148-4155. doi:
10.1002/cncr.27394 [doi].
105.	 Baco E, Gelet A, Crouzet S, et al. Hemi salvage high-intensity focused ultrasound (HIFU)
in unilateral radiorecurrent prostate cancer: A prospective two-centre study. BJU Int.
2014;114(4):532-540. doi: 10.1111/bju.12545 [doi].
106.	 Hsu CC, Hsu H, Pickett B, et al. Feasibility of MR imaging/MR spectroscopy-planned focal partial
salvage permanent prostate implant (PPI) for localised recurrence after initial PPI for prostate
cancer. Int J Radiat Oncol Biol Phys. 2013;85(2):370-377. doi: 10.1016/j.ijrobp.2012.04.028 [doi].
107.	 Peters M, Maenhout M, van der Voort van Zyp JRN, et al. Focal salvage iodine-125 brachytherapy
for prostate cancer recurrences after primary radiotherapy: A retrospective study regarding
toxicity, biochemical outcome and quality of life. Radiother Oncol. 2014;112(1):77-82. doi:
10.1016/j.radonc.2014.06.013 [doi].
108.	 Nguyen PL, Chen MH, D'Amico AV, et al. Magnetic resonance image-guided salvage brachytherapy
after radiation in select men who initially presented with favorable-risk prostate cancer: A
prospective phase 2 study. Cancer. 2007;110(7):1485-1492. doi: 10.1002/cncr.22934 [doi].
Introduction and outline
30
109.	 Crook JM, Potters L, Stock RG, Zelefsky MJ. Critical organ dosimetry in permanent seed prostate
brachytherapy: Defining the organs at risk. Brachytherapy. 2005;4(3):186-194. doi: S1538-
4721(05)00108-X [pii].
110.	 Davis BJ, Horwitz EM, Lee WR, et al. American brachytherapy society consensus guidelines for
transrectal ultrasound-guided permanent prostate brachytherapy. Brachytherapy. 2012;11(1):6-
19. doi: 10.1016/j.brachy.2011.07.005 [doi].
111.	 Price JG, Stone NN, Stock RG. Predictive factors and management of rectal bleeding side effects
following prostate cancer brachytherapy. Int J Radiat Oncol Biol Phys. 2013;86(5):842-847. doi:
10.1016/j.ijrobp.2013.04.033 [doi].
112.	 Hathout L, Folkert MR, Kollmeier MA, Yamada Y, Cohen GN, Zelefsky MJ. Dose to the bladder
neck is the most important predictor for acute and late toxicity after low-dose-rate prostate
brachytherapy: Implications for establishing new dose constraints for treatment planning. Int J
Radiat Oncol Biol Phys. 2014;90(2):312-319. doi: 10.1016/j.ijrobp.2014.06.031 [doi].
113.	 Salembier C, Lavagnini P, Nickers P, et al. Tumour and target volumes in permanent prostate
brachytherapy: A supplement to the ESTRO/EAU/EORTC recommendations on prostate
brachytherapy. Radiother Oncol. 2007;83(1):3-10. doi: S0167-8140(07)00028-X [pii].
114.	 Snyder KM, Stock RG, Hong SM, Lo YC, Stone NN. Defining the risk of developing grade 2
proctitis following 125I prostate brachytherapy using a rectal dose-volume histogram analysis. Int
J Radiat Oncol Biol Phys. 2001;50(2):335-341. doi: S0360-3016(01)01442-0 [pii].
115.	 Henriquez I, Sancho G, Hervas A, et al. Salvage brachytherapy in prostate local recurrence after
radiation therapy: Predicting factors for control and toxicity. Radiat Oncol. 2014;9:102-717X-9-
102. doi: 10.1186/1748-717X-9-102 [doi].
Max Peters1
Maaike R. Moman1
Henk G. van der Poel2
Henk Vergunst3
Igle Jan de Jong4
Peter L.M. Vijverberg5
Jan J. Battermann1
Simon Horenblas2
Marco van Vulpen1
1 Department of Radiation Oncology, University Medical Centre Utrecht, Utrecht, The Netherlands
2 Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
3 Department of Urology, Canisius-Wilhelmina Hospital, Nijmegen, The Netherlands
4 Department of Urology, University Medical Centre Groningen, Groningen, The Netherlands
5 Department of Urology, St. Antonius Hospital, Nieuwegein, The Netherlands
This work was published previously in: World Journal of Urology (2013) 31:403–409.
DOI 10.1007/s00345-012-0928-8
Patterns of outcome and toxicity after salvage
prostatectomy, salvage cryosurgery and salvage
Iodine-125 brachytherapy for prostate cancer
recurrences after radiation therapy:
a multi-centre experience and
literature comparison
Chapter 1
Chapter1
Chapter 1
34
Abstract
Purpose
Current salvage treatments for recurrent prostate cancer after primary radiation
therapy include radical prostatectomy, cryosurgery and brachytherapy. Because
toxicity and failure rates are considerable, salvage treatments are not commonly
performed. As most centres perform only one preferred salvage technique, the
literature only describes single-centre outcomes from a particular salvage technique
with a limited number of patients. In this overview, five high-volume Dutch centres
describe their toxicity and outcome data using different salvage techniques: salvage
radical prostatectomy, salvage Iodine-125 (I-125) brachytherapy and salvage
cryosurgery. This provides insight on how salvage is performed in clinical practice
in the Netherlands.
Methods
A total of 129 patients from five different centres in the Netherlands were
retrospectively analysed. Biochemical failure (BF) was defined as PSA >0.1 ng/ml
for the salvage prostatectomy group (n=44) and PSA nadir + 2.0 ng/ml (Phoenix
definition) for the salvage cryosurgery (n=54) and salvage I-125 brachytherapy
group (n=31). Toxicity was scored according to the Common Terminology Criteria
for Adverse events (CTCAE v3.0).
Results
BF occurred in 25 (81%) patients in the brachytherapy group after mean follow-up
of 29 (SD 24) months, in 29 patients (66%) patients in the prostatectomy group
after mean follow-up of 22 (SD 25) months and in 33 patients (61%) patients in the
cryosurgery group after mean follow-up of 14 (SD 11) months. Severe (≥grade 3)
genitourinary and gastrointestinal toxicity was observed in up to 30% of patients
in all three cohorts.
Conclusion
This overview shows clinical practice of prostate cancer salvage in the Netherlands.
Significant failure and toxicity rates are observed, regardless of salvage technique.
Patients should be selected with great care before offering these salvage treatment
strategies.
Chapter1
Outcomes of whole-gland salvage in the Netherlands
35
Introduction
Asignificantproportionofprostatecancerpatientstreatedwithprimaryradiotherapy
develops a recurrence. Estimations of risks exceeding 50% have been presented for
the highest risk groups1-4
. The recurrence risk is dependent on various risk factors
such as initial PSA level (iPSA), PSA doubling time (PSADT), Gleason score and PSA
nadir after initial treatment5
. A significant number of patients are thought to harbour
organ-confined disease6,7
. It seems local recurrences are particularly situated at the
position of the originally largest tumour, or index lesion8
. The currently dominantly
applied curative salvage treatments for these recurrences are salvage brachytherapy,
prostatectomyandcryosurgery.Unfortunately,thesesalvagetreatmentsareassociated
with high failure and severe toxicity rates9,10
. As most centres perform one preferred
salvage technique, the literature only describes single-centre outcomes with a limited
number of patients. In this overview, five high-volume Dutch centres describe their
outcomes using different salvage techniques. This provides a view on how salvage is
performed in clinical practice in the Netherlands.
Methods and materials
Treatment outcome and toxicity were recorded retrospectively for the salvage
treatments given: salvage radical prostatectomy (SRP), salvage cryosurgery (SCS)
and salvage Iodine-125 (I-125) implantation/brachytherapy (SBT). Primary
treatments were external beam radiation therapy or I-125 implantation. All men
showed PSA failure afterward, and recurrences were confirmed by transrectal
ultrasound (TRUS) guided biopsies. Furthermore, a bone scan and CT-scan were
made to exclude any possible distant or lymph node metastases. A PET-CT scan
was done in the more recent patients, but this imaging modality is not general
practice in the Netherlands. Patients were selected for salvage treatment based on a
life expectancy of at least 10 years, and all were counselled with respect to expected
toxicity and cancer control based on the experience of the institute.
Salvage procedures
All SRP procedures were performed at the Netherlands Cancer Institute-Antoni
van Leeuwenhoek Hospital (NKI-AVL), Amsterdam, via a perineal approach. Since
2003, 44 patients underwent SRP. A pelvic lymph node dissection was performed
before SRP in all patients, to exclude metastases. A median of 13 lymph nodes were
analysed.Theprocedureisdescribedelsewhereinmoredetail11
.Nonervesparingwas
attempted. Five patients received hormonal treatment ≤3 months prior to surgery.
SBT was performed by I-125 seed implantation at the University Medical Centre
Chapter1
Chapter 1
36
Utrecht (UMCU). Since 1993, 31 patients have been treated with permanent I-125
seed implants, via a transperineal route under ultrasound guidance. The standard
target dose was ≥145 Gy12
. The technique has been described previously13
. Only
patients with a prostate volume ≤50 cc were eligible for implantation. A total
of five patients received hormonal treatment three months prior to salvage in
order to decrease the prostatic volume. For the implants and the doses received
by the surrounding structures (rectum, bladder, urethra), the following criteria
were used: the prostatic V100 (the volume of the prostate that receives 100%
of the prescribed dose) ≥95%, the prostatic D90
(minimal dose to 90% of the
prostatic volume) ≥145 Gy, the rectal D2cc (the minimal dose received by the
maximally irradiated 2cc of the structure) ≤145 Gy and the D10 (the minimal
dose to 10% of the structure) of the prostatic urethra ≤150% of 145 Gy12,14
.
Cryosurgery was performed at the Canisius-Wilhelmina Hospital (CWH),
Nijmegen, the University Medical Centre Groningen (UMCG) and the Sint
Antonius Hospital (St.AH), Nieuwegein. Since 2007, 54 patients underwent salvage
cryosurgery. Cryosurgery was performed with Seednet (prostate <35 cc) and
Icerod needles (prostate >35 cc) inserted transperineally through a brachytherapy
template. The 17 Gauge needles were placed 1 cm apart with TRUS visualisation. A
transurethral warming catheter was inserted to protect the urethra. A suprapubic
catheter was left in situ for two weeks, in combination with antibiotic prophylaxis.
Patients were treated with two cycles of freezing and thawing. Thermosensors in the
midportion of the prostate gland ensured that the required temperature of -40 0C
was reached for effective cell killing.
Biochemical failure and toxicity
BF for patients after SRP was defined as a serum PSA level >0.1 ng/ml. For SBT
and SCS, failure according to the Phoenix and ASTRO definition was assessed:
PSA nadir + 2 ng/ml (Phoenix) and three consecutive rises in PSA with backdating
of the moment of failure to midway between the nadir after treatment and the
first rise (ASTRO)15,16
. Toxicity was scored from file records based on structured
interviewsatdifferenttimeintervalsaftersalvagetreatment.Toimproveuniformity,
the toxicity from each clinic was retrospectively converted to a grade using the
Common Terminology Criteria for Adverse Events, version 3.0 (CTCAE v3.0)17
.
No subdivision into acute and late toxicity was made for this analysis.
Statistical analysis
Descriptive statistics and Kaplan–Meier survival analyses for freedom from
biochemical failure (FFF) were performed with the Statistical Package for the
Chapter1
Outcomes of whole-gland salvage in the Netherlands
37
Social Sciences, version 17.0 (SPSS, Chicago, IL, USA). Differences in Kaplan-
Meier FFF estimates were tested with the log-rank test, with statistical significance
set at p ≤0.05. Toxicity is presented as the proportion of patients suffering from
genitourinary (GU) or gastrointestinal (GI) morbidity and erectile dysfunction
(ED).
Results
A total of 129 patients were included. Various baseline characteristics differed
betweenthegroups,inparticularage,iPSA,clinicaltumourstageandfollow-uptime
(Table 1). SBT and SCS patients were older at the time of salvage than SRP patients
(69 and 70 years vs. 65 years). The iPSA was higher than 20 ng/ml in 35% of the SBT
group and 30% of the SCS group, versus 5% in the SRP group. Primary cT3 tumours
were19%inSBTpatients,32%inSRPpatientsand39%inSCSpatients.Meanfollow-
up in the SCS group was 3 years, in comparison with 5 and 9 years for SRP and SBT,
respectively.Resultsregardingbiochemicalresponse,mortalityandsevere(grade3)
toxicity are presented in table 2. Figure 1 shows the Kaplan–Meier curve for the FFF.
In the SRP group, 29 (66%) patients suffered from BF after mean 22 months (SD
25 months). Grade 1 or 2 GU toxicity occurred in 24 (55%) patients, and grade 3
toxicity occurred in 10 (23%) patients. Grade 1 or 2 GI toxicity occurred in three
(7%) patients, and grade 3 (fistulas) occurred in four (9%) patients. Until now, five
(11%)patientshavediedduetoprostatecancer.EDwasobservedin86%ofpatients.
After I-125 implantation, 25 (81%) patients suffered from BF after mean 29 months
(SD 24 months). Grade 1 or 2 GU toxicity occurred in 17 (55%) patients, and grade
3 occurred in seven (23%) patients, predominantly urinary incontinence and
obstruction due to swelling of the prostate. Grade 1 and 2 GI toxicity occurred
in 16 (52%) patients, and grade 3 occurred in two (6%) patients. Twelve (39%)
patients have died due to prostate cancer. ED was present in 45% of patients.
After a mean 14 months after SCS, 33 (61%) patients suffered from BF after mean 14
months (SD 11 months). Grade 1 and 2 GU toxicity occurred in 30 (56%) patients,
and grade 3 occurred in 12 (22%) patients. Urinary incontinence, obstruction
and urinary tract infection were frequent complications. Perineal pain was also a
common complaint. Grade 1 and 2 GI toxicity occurred in seven (13%) patients,
and grade 3 occurred in four (7%) patients (predominantly fistula formation
between the rectum and urethra). ED was observed in 50 (93%) patients. Twenty-
four (44%) patients had pre-existent ED. No patients have died due to prostate
cancer–related events.
Chapter1
Chapter 1
38
Table 1: Baseline characteristics of the study population, per salvage treatment group
Prostatectomy I-125 Cryosurgery
N 44 31 54
Mean (SD) age at primary diagnosis, years 60 (6) 64 (5) 68 (6)
Mean (SD) age at salvage treatment, years 65 (6) 69 (5) 70 (6)
Primary tumour stage
cT1
cT2
cT3
10 (23%)
20 (46%)
14 (32%)
5 (16%)
20 (65%)
6 (19%)
16 (30%)
17 (31%)
21 (39%)
Gleason score
Gleason 2 - 6
Gleason 7
Gleason 8 – 10
Unknown
18 (41%)
19 (43%)
7 (16%)
13 (42%)
13 (42%)
2 (6%)
3 (10%)
14 (26%)
16 (30%)
21 (39%)
3 (5%)
Primary treatment
EBRT
HDR
I-125
IMRT
31 (70%)
2 (5%)
11 (25%)
0
20 (64%)
0
11 (36%)
0
43 (80%)
0
4 (7%)
7 (13)
Initial PSA
0 – 10 ng/ml
> 10 – 20 ng/ml
> 20 ng/ml
Unknown
12 (27%)
29 (66%)
2 (5%)
1 (2%)
5 (16%)
7 (23%)
11 (35%)
8 (26%)
16 (30%)
15 (27%)
16 (30%)
7 (13%)
PSA before salvage, ng/ml
0 – 10 ng/ml
> 10 – 20 ng/ml
> 20 ng/ml
Unknown
24 (55%)
18 (41%)
2 (5%)
17 (55%)
8 (26%)
5 (16%)
1 (3%)
42 (78%)
9 (17%)
3 (5%)
0
Mean (SD) PSADT before salvage, months 13 (8) 13 (9) 15 (5)
Mean (SD) prostate volume before salvage, cm3
28 (2) 24 (10) 21 (8)
Number of patients treated with hormonal
therapy <3 months before salvage
5 (11%) 5 (16%) 8 (15%)
Abbreviations: I-125=Iodine-125; SD=standard deviation; EBRT=external beam radiotherapy;
HDR=high dose rate radiotherapy; IMRT=Intensity Modulated Radiotherapy; PSADT=PSA
doubling time.
Chapter1
Outcomes of whole-gland salvage in the Netherlands
39
Table 2: Biochemical failure, mortality and toxicity rates after salvage, per salvage treatment group
Prostatectomy I-125 Cryosurgery
N 44 31 54
Mean (SD) FU after salvage, years 5 (4) 9 (3) 3 (0,3)
Mean (SD) PSA nadir, ng/ml 0.2 (0.8) 4.7 (6.6) 2 (3)
Mean (SD) time to nadir, months 1.5 10 (9) 2 (2)
Patients with biochemical failure†
Phoenix
ASTRO
PSA >0.1 ng/ml
NA
NA
29 (66%)
25 (81%)
17 (55%)
NA
33 (61%)
32 (59%)
NA
Mean (SD) time to failure, months
Phoenix
ASTRO
PSA >0.1 ng/ml
NA
NA
22 (25)
29 (24)
41 (25)
NA
14 (11)
16 (9)
NA
Patients (%) with severe GU toxicity, CTCAE
Grade 3 10 (23%) 7 (23%) 12 (22%)
Patients (%) with severe GI toxicity, CTCAE
Grade 3 4 (9%) 2 (6%) 4 (7%)
Death
Yes, PCa-specific
Yes, other cause
5 (11%)
7 (16%)
12 (39%)
2 (6%)
0 (0%)
5 (9%)
Abbreviations: I-125=Iodine-125; FU=follow up; SD=standard deviation; ASTRO=American
Society for Therapeutic Radiology and Oncology; GU=genitourinary; GI=gastrointestinal;
PCa=prostate cancer; CTCAE=Common Terminology Criteria for Adverse Events v3.0; NA=Not
applicable.
†
Proportion of patients with biochemical failure, as defined by 2 different criteria. Definitions of
biochemical failure: Phoenix=PSA nadir plus 2 ng/ml, ASTRO=3 consecutive rises in PSA backdating
the moment of failure to midway between the nadir after treatment and the first rise.
Chapter1
Chapter 1
40
Discussion
This paper shows how salvage for radiorecurrent prostate cancer is performed in
the Netherland and reports on the current cancer control outcomes and toxicity.
Biochemical failure occurs often, with an estimated FFF of maximally 27% after
5years.Toxicitywasalsofrequent,withapproximately30%ofpatientsexperiencing
a grade 3 or higher complication of the genitourinary and gastrointestinal tract
combined.Intheliterature,awidevariationofpapersdescribestreatmentoutcomes
and toxicity after different salvage modalities, preceded by different primary
radiation therapy modalities and schedules. To our knowledge, no previous article
has presented outcomes of three different salvage treatment modalities from one
population. A comparison with the literature is summarised in table 3 (FFF) and
table 4 (toxicity).
Figure 1: Kaplan-Meier curve showing the freedom from biochemical failure following salvage radical
prostatectomy, salvage Iodine-125 implantation and salvage cryotherapy (Log rank: p=0.42). The
numbers at risk at 5 years were 7 for salvage radical prostatectomy, 5 for Iodine-125 brachytherapy and
6 (at three years) for salvage cryosurgery.
Chapter1
Outcomes of whole-gland salvage in the Netherlands
41
Table 3: Comparison with results from the literature – freedom from biochemical failure.
Salvage procedure No. studies
[References]
N Follow up FFF
(%)
Follow up
present study
FFF present
study (%)
Radical prostatectomy 4[10,11,19,27,28]
337 5 years 31 – 71 5 years 27
I-125 implantation 2[10,21,29]
66 5 years 34 – 53 5 years 19
Cryosurgery 3[10,22,24,30]
597 5 years 40 – 55 3 years 19
Abbreviations: FFF=freedom from biochemical failure; I-125=Iodine-125.
Table 4: Comparison with results from the literature – toxicity
Salvage procedure Radical prostatectomy I-125 implantation Cryosurgery
Literature
No. studies
[References]
4[10,11,19,27,31]
2[10,21,29]
2[10,23,30]
N 335 66 397
% GU toxicity 20-68% incontinence
22-41% bladder neck
stricture
0-47% grade 3-4 4-83% incontinence
7-55% bladder neck
stricture / retention
% GI toxicity 2-7% rectal injury 0-24% grade 3-4 6-37% perineal pain
1-11% fistula
Erectile dysfunction 72 %—Nearly uniform NA 72–86 %
Present study
% GU toxicity 23% grade 3 23% grade 3 22% grade 3
% GI toxicity 9% grade 3 6% grade 3 7% grade 3
Erectile dysfunction 86% 45% 93%*
Abbreviations: I-125=Iodine-125; GU=genitourinary; GI=gastrointestinal. NA=Not available
* 44% of patients had pre-existent erectile dysfunction.
However,thiscomparisonishamperedbythesmallanddivergentstudypopulations
and the different definitions for BF. Although the current study population is not
equivalent regarding patient and tumour characteristics and follow-up, it might be
possible to make a crude comparison regarding toxicity. Grade 3 GU and GI toxicity
occurredin32%ofSRPpatients,29%ofSBTpatientsand30%ofSCSpatients.These
toxicity rates are approximately equivalent to results as found in the literature9,10
.
The5-yearFFFwas27%followingSRPand19%followingSBT(Phoenixdefinition).
After 36 months, the FFF following cryosurgery was 19%. These figures are lower
Chapter1
Chapter 1
42
compared to those found in the literature (Table 3 & 4 and figure 1). This could be
due to incomplete salvage procedures, but dissemination before salvage is more
likely. Especially the cryosurgery group contained high risk cases with cT3 and
Gleason8–10tumours.Theliteratureshowsthatthepre-salvagePSAconcentration,
PSADT after primary radiation treatment and the tumour stage and grade are
important in predicting biochemical recurrences after salvage procedures9-11,18
. In
one SRP study, patients with a pre-salvage PSA <2 ng/ml and PSADT >12 months
showed better biochemical disease free survival (bDFS) rates than patient with
PSA >2 ng/ml and PSADT <12 months: >80% bDFS after 80 months in the most
favourable risk group compared with 0% bDFS after 20 months in the group with
the highest risk factors11
. Other SRP data indicate more favourable results for
primary T2 tumours in comparison with T3 tumours (100% bDFS after 5 years
in T2 tumours compared to 35% in T3 and 0% in N+ tumours)19
. In addition, the
tumour stage (T3/T4) was found to be an independent predictor of cancer specific
survival in a large series of SRP patients after multivariable analysis (n=199)18
.
In SBT, patients with pre-salvage Gleason scores ≥7 and PSADT <10 months show
poorer biochemical control rates in multivariable analysis10,20
. Possibly, a pre-
salvage PSA >10 ng/ml also leads to poorer cancer control outcomes21
. For the SCS
group, these risk factors for salvage treatment failure have also been recognised.
Patients with a pre-salvage PSA >10 ng/ml, Gleason scores ≥8 before primary
treatment and clinical T3/T4 tumours show decreased biochemical control rates
in comparison with patients without these risk factors9,10,22-24
. For an accurate
selection of patients likely to benefit from salvage therapy, these risk factors have to
be incorporated in salvage protocols. The general absence of these selection criteria
in these cohorts has probably contributed to the fairly low cancer control rates
observed. These pre-salvage characteristics would need to be researched with the
use of adequate prediction modelling to aid patient selection in the future.
The retrospective nature of this analysis yields several additional limitations.
First, the study populations are different at baseline, and therefore especially the
results regarding cancer control are difficult to compare between the three salvage
modalities. Also, the FFF rates depend on the criteria used to define BF. For SBT and
SCS, the Phoenix definition was used, because the ASTRO definition is more prone
to bias because of variable follow-up intervals. The ASTRO definition furthermore
has a false-positive rate of approximately 20% after the use of hormonal therapy,
versus5%associatedwiththePhoenixdefinition15,16
.Further,becausethegroupsare
limited, analysis of risk factors in a multivariable model was not perfomed. Finally,
the use of hormonal treatment was not standardised as well, further hampering
Chapter1
Outcomes of whole-gland salvage in the Netherlands
43
comparisons between groups. The primary objective of this study, however, was not
to provide an exhaustive comparison regarding different salvage modalities, but to
show the current clinical practice in the Netherlands. This can provide an indication
of what aspects need to be considered when offering salvage. Patients should be
intensely counselled regarding the high toxicity rates. These numbers might even
be underestimated due to the retrospective nature of the analysis, creating the
possibilitythatsalvagecanbeevenmoredetrimentalthanobservedinthesecohorts.
In addition, cancer control outcomes are suboptimal, which is possibly related to
inaccurate patient selection. In the SBT group, treatment goes back as far as 1993.
The patients selected in this period often had high-risk characteristics that would
exclude them from salvage treatment today. It is possible that especially this lack of
proper patient selection has negatively influenced the FFF. For future research, it is
important to pay further attention to patient selection. The most important aspect is
clearly defining local or distant disease at the time of BF after primary radiotherapy.
The PSA value is one of the most important diagnostic and follow-up measurement
available today, but is insufficient to assess disease progression. A rising PSA level
after primary radiotherapy can have several possible origins, including lymph-node
or distant metastases. Furthermore, the currently applied Phoenix definition has
a false positive rate of 5%16
. Better imaging techniques could contribute to a better
selection of patients with a local (organ-confined) recurrence and exclude patients
with systemic disease25,26
. Prediction models taking into account several pre-salvage
characteristicscouldfurtheraidpatientselection.Thesemodelsincombinationwith
accurate diagnostic modalities should be able to lead to a better patient selection
for salvage treatment in the future, thereby improving cancer control outcomes.
Lastly, with better assessment of localised disease, focal salvage directed only at
the recurrent tumour area might be able to decrease the often high toxicity rates
associated with a whole-gland approach by reducing cumulative damage to
previously irradiated surrounding organs at risk.
Conclusion
Both biochemical failure and toxicity rates are considerable after salvage radical
prostatectomy, Iodine-125 brachytherapy and cryosurgery. Improvements in
patient selection using pre-salvage characteristics and other diagnostic modalities
can possibly contribute to better outcomes in the future.
Chapter1
Chapter 1
44
References
1.	 Agarwal PK, Sadetsky N, Konety BR, Resnick MI, Carroll PR, Cancer of the Prostate Strategic
Urological Research Endeavor (CaPSURE). Treatment failure after primary and salvage therapy
for prostate cancer: Likelihood, patterns of care, and outcomes. Cancer. 2008;112(2):307-314. doi:
10.1002/cncr.23161.
2.	 Heidenreich A, Aus G, Bolla M, et al. EAU guidelines on prostate cancer. Eur Urol. 2008;53(1):68-
80. doi: 10.1016/j.eururo.2007.09.002.
3.	 Kuban DA, Thames HD, Levy LB, et al. Long-term multi-institutional analysis of stage T1-
T2 prostate cancer treated with radiotherapy in the PSA era. Int J Radiat Oncol Biol Phys.
2003;57(4):915-928.
4.	 Hinnen KA, Battermann JJ, van Roermund JG, et al. Long-term biochemical and survival outcome
of 921 patients treated with I-125 permanent prostate brachytherapy. Int J Radiat Oncol Biol Phys.
2010;76(5):1433-1438. doi: 10.1016/j.ijrobp.2009.03.049.
5.	 Zelefsky MJ, Pei X, Chou JF, et al. Dose escalation for prostate cancer radiotherapy: Predictors of
long-term biochemical tumour control and distant metastases-free survival outcomes. Eur Urol.
2011;60(6):1133-1139. doi: 10.1016/j.eururo.2011.08.029 [doi].
6. 	 Ahmed HU, Pendse D, Illing R, Allen C, van der Meulen JH, Emberton M. Will focal therapy
become a standard of care for men with localised prostate cancer? Nat Clin Pract Oncol.
2007;4(11):632-642. doi: 10.1038/ncponc0959.
7. 	 Pound CR, Brawer MK, Partin AW. Evaluation and treatment of men with biochemical prostate-
specific antigen recurrence following definitive therapy for clinically localised prostate cancer. Rev
Urol. 2001;3(2):72-84.
8. 	 Pucar D, Hricak H, Shukla-Dave A, et al. Clinically significant prostate cancer local recurrence
after radiation therapy occurs at the site of primary tumour: Magnetic resonance imaging and
step-section pathology evidence. Int J Radiat Oncol Biol Phys. 2007;69(1):62-69. doi: 10.1016/j.
ijrobp.2007.03.065.
9.	 Kimura M, Mouraviev V, Tsivian M, Mayes JM, Satoh T, Polascik TJ. Current salvage methods for
recurrent prostate cancer after failure of primary radiotherapy. BJU Int. 2010;105(2):191-201. doi:
10.1111/j.1464-410X.2009.08715.x.
10.	 Nguyen PL, D'Amico AV, Lee AK, Suh WW. Patient selection, cancer control, and complications
after salvage local therapy for postradiation prostate-specific antigen failure: A systematic review
of the literature. Cancer. 2007;110(7):1417-1428. doi: 10.1002/cncr.22941.
11.	 van der Poel HG, Beetsma DB, van Boven H, Horenblas S. Perineal salvage prostatectomy for
radiation resistant prostate cancer. Eur Urol. 2007;51(6):1565-71; discussion 1572. doi: 10.1016/j.
eururo.2006.08.011.
12.	 Ash D, Flynn A, Battermann J, et al. ESTRO/EAU/EORTC recommendations on permanent seed
implantation for localised prostate cancer. Radiother Oncol. 2000;57(3):315-321.
Chapter1
Outcomes of whole-gland salvage in the Netherlands
45
13.	 Battermann JJ. I-125 implantation for localised prostate cancer: The utrecht university experience.
Radiother Oncol. 2000;57(3):269-272.
14.	 Salembier C, Lavagnini P, Nickers P, et al. Tumour and target volumes in permanent prostate
brachytherapy: A supplement to the ESTRO/EAU/EORTC recommendations on prostate
brachytherapy. Radiother Oncol. 2007;83(1):3-10. doi: S0167-8140(07)00028-X [pii].
15.	 RoachM,3rd,HanksG,ThamesH,Jr,etal.Definingbiochemicalfailurefollowingradiotherapywith
or without hormonal therapy in men with clinically localised prostate cancer: Recommendations
of the RTOG-ASTRO phoenix consensus conference. Int J Radiat Oncol Biol Phys. 2006;65(4):965-
974. doi: 10.1016/j.ijrobp.2006.04.029.
16.	 Buyyounouski MK, Hanlon AL, Eisenberg DF, et al. Defining biochemical failure after
radiotherapy with and without androgen deprivation for prostate cancer. Int J Radiat Oncol Biol
Phys. 2005;63(5):1455-1462. doi: 10.1016/j.ijrobp.2005.05.053.
17.	 Trotti A, Colevas AD, Setser A, et al. CTCAE v3.0: Development of a comprehensive grading
system for the adverse effects of cancer treatment. Semin Radiat Oncol. 2003;13(3):176-181. doi:
10.1016/S1053-4296(03)00031-6.
18. 	 Ward JF, Pagliaro LC, Pisters LL. Salvage therapy for radiorecurrent prostate cancer. Curr Probl
Cancer. 2008;32(6):242-271. doi: 10.1016/j.currproblcancer.2008.10.001.
19. 	 Sanderson KM, Penson DF, Cai J, et al. Salvage radical prostatectomy: Quality of life outcomes
and long-term oncological control of radiorecurrent prostate cancer. J Urol. 2006;176(5):2025-31;
discussion 2031-2. doi: S0022-5347(06)01767-8 [pii].
20. 	 Moman MR, van der Poel HG, Battermann JJ, Moerland MA, van Vulpen M. Treatment outcome
and toxicity after salvage 125-I implantation for prostate cancer recurrences after primary 125-I
implantation and external beam radiotherapy. Brachytherapy. 2010;9(2):119-125. doi: 10.1016/j.
brachy.2009.06.007.
21. 	 Beyer DC. Permanent brachytherapy as salvage treatment for recurrent prostate cancer. Urology.
1999;54(5):880-883.
22. 	 Ng CK, Moussa M, Downey DB, Chin JL. Salvage cryoablation of the prostate: Followup and
analysis of predictive factors for outcome. J Urol. 2007;178(4 Pt 1):1253-7; discussion 1257. doi:
10.1016/j.juro.2007.05.137.
23. 	 Chin JL, Pautler SE, Mouraviev V, Touma N, Moore K, Downey DB. Results of salvage cryoablation
of the prostate after radiation: Identifying predictors of treatment failure and complications. J Urol.
2001;165(6 Pt 1):1937-41; discussion 1941-2.
24. 	 Izawa JI, Madsen LT, Scott SM, et al. Salvage cryotherapy for recurrent prostate cancer after
radiotherapy: Variables affecting patient outcome. J Clin Oncol. 2002;20(11):2664-2671.
25. 	 Haider MA, Chung P, Sweet J, et al. Dynamic contrast-enhanced magnetic resonance imaging for
localisation of recurrent prostate cancer after external beam radiotherapy. Int J Radiat Oncol Biol
Phys. 2008;70(2):425-430. doi: 10.1016/j.ijrobp.2007.06.029.
Chapter1
Chapter 1
46
26. 	 Barentsz JO, Richenberg J, Clements R, et al. ESUR prostate MR guidelines 2012. Eur Radiol.
2012;22(4):746-757. doi: 10.1007/s00330-011-2377-y; 10.1007/s00330-011-2377-y.
27. 	 Ward JF, Sebo TJ, Blute ML, Zincke H. Salvage surgery for radiorecurrent prostate cancer:
Contemporary outcomes. J Urol. 2005;173(4):1156-1160. doi: 10.1097/01.ju.0000155534.54711.60.
28. 	 Bianco FJ,Jr, Scardino PT, Stephenson AJ, Diblasio CJ, Fearn PA, Eastham JA. Long-term oncologic
results of salvage radical prostatectomy for locally recurrent prostate cancer after radiotherapy. Int
J Radiat Oncol Biol Phys. 2005;62(2):448-453. doi: 10.1016/j.ijrobp.2004.09.049.
29. 	 Grado GL, Collins JM, Kriegshauser JS, et al. Salvage brachytherapy for localised prostate cancer
after radiotherapy failure. Urology. 1999;53(1):2-10.
30. 	 PistersLL,RewcastleJC,DonnellyBJ,LugnaniFM,KatzAE,JonesJS.Salvageprostatecryoablation:
Initial results from the cryo on-line data registry. J Urol. 2008;180(2):559-63; discussion 563-4.
doi: 10.1016/j.juro.2008.04.005.
31. 	 Stephenson AJ, Scardino PT, Bianco FJ,Jr, DiBlasio CJ, Fearn PA, Eastham JA. Morbidity and
functional outcomes of salvage radical prostatectomy for locally recurrent prostate cancer after
radiation therapy. J Urol. 2004;172(6 Pt 1):2239-2243.
 
p
Daniël A. Smit Duijzentkunst*1
Max Peters*1
Jochem R.N. van der Voort van Zyp1
Marinus A. Moerland1
Marco van Vulpen1
1 Department of Radiation Oncology, University Medical Centre Utrecht, The Netherlands
* Both authors contributed equally to this research
This chapter requires revisions for: World Journal of Urology
Chapter 2
Focal salvage therapy for
local prostate cancer recurrences
after primary radiotherapy:
a comprehensive review
Chapter2
Chapter 2
50
Abstract
Background/Aim
Patients with locally recurrent prostate cancer after primary radiotherapy can be
eligible for salvage treatment. Whole-gland salvage techniques carry a high risk
of toxicity. A focal salvage approach might reduce the risk of adverse events while
maintaining cancer control in carefully selected patients. The aim of this review is
to evaluate current literature to assess whether focal salvage leads to a comparable
or favourable recurrence rate and less toxicity compared to whole-gland salvage.
Methods
A literature search was performed using PubMed, Embase and the Cochrane
Library. 3015 Articles were screened and assessed for quality. Eight papers (on
focal cryoablation (n=3), brachytherapy (n=3) and HIFU (n=2)) were used to
report outcomes.
Results
1, 2, 3 and 5 year biochemical disease free survival (BDFS) ranges for focal salvage
are respectively 69%-100%, 49%-100%, 50%-91% and 46.5%-54.5%. Severe
genitourinary, gastrointestinal and sexual function toxicity rates are 0%-33.3%.
One study directly compares focal to whole-gland salvage cryotherapy, showing
5-year BDFS of respectively 54.4% and 86.5% with lower toxicity rates for focal
salvage patients.
Conclusion
Provisional data suggest that BDFS rates of focal salvage are in line with those of
whole-gland approaches. There is evidence that focal salvage could decrease severe
toxicity and preserve erectile function.
Focal salvage for radiorecurrent prostate cancer – a review
Chapter2
51
Introduction
Prostate cancer (PCa) patients primarily treated with external beam radiotherapy
(EBRT) or brachytherapy (BT) are at risk of a recurrence, depending on their
pre-treatment characteristics. The intermediate and high risk groups can
suffer from a biochemical recurrence in over 50% of the cases after 10 years
follow-up1,2
. Subsequently, these biochemical recurrences give rise to a risk
of developing metastases and (PCa specific) mortality3
. Up to 98% of patients
receive (palliative) androgen deprivation therapy (ADT) as treatment after a
biochemical recurrence4
. However, a substantial amount of patients harbour
organ-confined disease, for which a curative salvage procedure can provide
durable disease control5,6
, without exposing patients to the often severe side-
effects of ADT7
. Salvage nowadays is usually performed using a whole-gland
approach, which is accompanied by a high chance of severe gastro-intestinal (GI),
genitourinary (GU) and erectile toxicity due to previous radiation damage to
surrounding organs at risk5,6
. Given the evidence from pathology studies that
recurrences are frequently localised at the site of the primary largest (index)
tumour8,9
, a focal salvage approach, directed solely at the area containing
recurrent tumour, might be a viable treatment option for patients with unifocal
PCa recurrences without metastatic disease. This way, the serious adverse events
associated with whole-gland salvage might be prevented, while cancer control is
maintained. The current literature regarding survival and functional outcomes of
focal salvage techniques for prostate cancer recurrences after primary radiotherapy
is evaluated here.
Materials and methods
Literature search
On 19-08-2015 a systematic literature search was performed in the Pubmed,
Embase and Cochrane Library databases. In order to create a sensitive search, the
search syntax was build based on domain and determinant (Table 1). After removal
of duplicates, 3015 papers were screened, selecting 55 articles with matching
domain and determinant. Exclusion criteria are listed in the flowchart (Figure 1).
An additional reference search was performed, resulting in 12 studies for this
review.
Study selection
Studies were subjected to a critical appraisal based on an adaptation of the Dutch
Cochrane Centre recommendations (ACROBAT-NRSI risk of bias assessment
Chapter2
Chapter 2
52
Table 1: Search Syntax    
All search terms (title/abstract) for Pubmed, (ab,ti) for Embase, (ti,ab,kw) for Cochrane
1 Salvage 14 Local
2 Therapy 15 Focal
3 Treatment 16 Prostate
4 Rescue 17 Prostatic
5 Cryoablation 18 Cancer
6 Cryosurgery 19 Carcinoma
7 Cryotherapy 20 Adenocarcinoma
8 Ablation 21 Neoplasma
9 Brachytherapy 22 Recurrence
10 HIFU 23 Recurrences
11 "high intensity focused ultrasound" 24 Recurrent
12 Hemi 25 Relapse
13 Partial 26 Radiorecurrent
       
27 1 or 2 or 3 or 4 or 5 or 6 or 7 or 8 or 9 or 10 or 11
28 12 or 13 or 14 or 15    
29 27 and 28    
30 16 or 17 #34 Search Results
31 18 or 19 or 20 or 21 Pubmed: 1719
32 22 or 23 or 24 or 25 or 26 Embase: 2811
33 30 and 31 and 32 Cochrane: 199
34 29 and 33 Total: 4729
tool)10
. Studies were graded (+), (+) or (-) on relevance (domain, determinant,
outcome) and validity (selection, study population characteristics, exposure,
primary outcome, secondary outcomes, follow-up and number of patients) (Table
A1 (supplementary file)).
All studies were observational cohorts or case series with either prospective or
retrospective data acquisition. In that aspect, no study could be considered of high
quality. In none of the studies blinding was applied. However, the primary outcome
(BDFS) is unlikely to be influenced by information bias, due to the objectivity of
the failure definition (Phoenix, i.e. PSA-nadir + 2.0 ng/ml). On the contrary, the
secondary outcome is subjective to information bias and is graded in that way.
Confounding was not graded, since all studies except one were single-arm
Focal salvage for radiorecurrent prostate cancer – a review
Chapter2
53
4729
3015
55
11
12
Embase
2811
Pubmed
1719
Removal
of duplicates
Inclusion criteria
Domain = locally
recurrent PCa after
primary radiotherapy
Determinant = focal
salvage treatment
Exclusion criteria
No full text = 6
Review = 10
Conference abstract/
poster = 19
Comment/opinion/
reply = 2
Study protocol = 1
Guideline = 1
Technical report = 2
Reprinted article = 3
Screening
title/abstract
Full text
evaluation
Related
articles
Cochrane
199
Figure 1. Flowchart
observational cohorts. The study by De Castro et al.11
describes a two-armed cohort.
Here, possible confounding factors are adequately described, but not corrected for
in multivariable analysis. Four studies describe case series with 10 patients or less.
These were valued as low quality of evidence and therefore excluded.
Results
Search results
A total of 8 studies were eligible for data extraction, describing focal salvage
treatment with cryoablation (CA) (n=3), low-dose-rate brachytherapy (LDR-
BT) (n=3) and high intensity focused ultrasound (HIFU) (n=2) in a total of 278
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters
Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters

More Related Content

What's hot

HCC: clinical update AASLD based
HCC: clinical update AASLD basedHCC: clinical update AASLD based
HCC: clinical update AASLD basedAhmed Elmoughazy
 
ECCLU 2011 - N. James - Localised invasive bladder cancer - Radiotherapy
ECCLU 2011 - N. James - Localised invasive bladder cancer - RadiotherapyECCLU 2011 - N. James - Localised invasive bladder cancer - Radiotherapy
ECCLU 2011 - N. James - Localised invasive bladder cancer - RadiotherapyEuropean School of Oncology
 
Radiotherapy for bladder cancers
Radiotherapy for bladder cancersRadiotherapy for bladder cancers
Radiotherapy for bladder cancersAshutosh Mukherji
 
Radiotherapy Planning For Esophageal Cancers
Radiotherapy Planning For Esophageal CancersRadiotherapy Planning For Esophageal Cancers
Radiotherapy Planning For Esophageal Cancersfondas vakalis
 
Cytoreductive nephrectomy (when and why)
Cytoreductive nephrectomy (when and why)Cytoreductive nephrectomy (when and why)
Cytoreductive nephrectomy (when and why)Ahmad Kharrouby
 
What Increases Penile Growth Kit
What Increases Penile Growth KitWhat Increases Penile Growth Kit
What Increases Penile Growth KitRaven Butler
 
Personalised medicine in rt dr. ashutosh
Personalised medicine in rt   dr. ashutoshPersonalised medicine in rt   dr. ashutosh
Personalised medicine in rt dr. ashutoshAshutosh Mukherji
 
ECCLU 2011 - S. Supiot - Locally advanced prostate cancer: Multimodality trea...
ECCLU 2011 - S. Supiot - Locally advanced prostate cancer: Multimodality trea...ECCLU 2011 - S. Supiot - Locally advanced prostate cancer: Multimodality trea...
ECCLU 2011 - S. Supiot - Locally advanced prostate cancer: Multimodality trea...European School of Oncology
 
Prostate cancer updates 2021
Prostate cancer updates 2021Prostate cancer updates 2021
Prostate cancer updates 2021Kanhu Charan
 
LUNG CANCER MANAGEMENT IN LOW RESOURCE SETTINGS
LUNG CANCER MANAGEMENT IN LOW RESOURCE SETTINGSLUNG CANCER MANAGEMENT IN LOW RESOURCE SETTINGS
LUNG CANCER MANAGEMENT IN LOW RESOURCE SETTINGSKanhu Charan
 
ECCLU 2011 - A. Widmark - Prostate cancer: All the truth about local treatmen...
ECCLU 2011 - A. Widmark - Prostate cancer: All the truth about local treatmen...ECCLU 2011 - A. Widmark - Prostate cancer: All the truth about local treatmen...
ECCLU 2011 - A. Widmark - Prostate cancer: All the truth about local treatmen...European School of Oncology
 
Adjuvant radiation based on genomic risk factors emerging scenarios
Adjuvant radiation based on genomic risk factors   emerging scenariosAdjuvant radiation based on genomic risk factors   emerging scenarios
Adjuvant radiation based on genomic risk factors emerging scenariosSantam Chakraborty
 
ROLE OF RADIATION IN BONE TUMORS FOR ORTHOPEDICS
ROLE OF RADIATION IN BONE TUMORS FOR ORTHOPEDICSROLE OF RADIATION IN BONE TUMORS FOR ORTHOPEDICS
ROLE OF RADIATION IN BONE TUMORS FOR ORTHOPEDICSKanhu Charan
 
Results of Stereotactic Body Radiotherapy (SBRT) for Management of Hepatic Tu...
Results of Stereotactic Body Radiotherapy (SBRT) for Management of Hepatic Tu...Results of Stereotactic Body Radiotherapy (SBRT) for Management of Hepatic Tu...
Results of Stereotactic Body Radiotherapy (SBRT) for Management of Hepatic Tu...Premier Publishers
 
ECCLU 2011 - M. Bolla - Prostate cancer: Locally advanced disease and patient...
ECCLU 2011 - M. Bolla - Prostate cancer: Locally advanced disease and patient...ECCLU 2011 - M. Bolla - Prostate cancer: Locally advanced disease and patient...
ECCLU 2011 - M. Bolla - Prostate cancer: Locally advanced disease and patient...European School of Oncology
 
SBRT in head and neck cancer
SBRT in  head and neck cancerSBRT in  head and neck cancer
SBRT in head and neck cancerDr Rushi Panchal
 

What's hot (20)

HCC: clinical update AASLD based
HCC: clinical update AASLD basedHCC: clinical update AASLD based
HCC: clinical update AASLD based
 
ECCLU 2011 - N. James - Localised invasive bladder cancer - Radiotherapy
ECCLU 2011 - N. James - Localised invasive bladder cancer - RadiotherapyECCLU 2011 - N. James - Localised invasive bladder cancer - Radiotherapy
ECCLU 2011 - N. James - Localised invasive bladder cancer - Radiotherapy
 
IMRT in Prostate Cancer
IMRT in Prostate CancerIMRT in Prostate Cancer
IMRT in Prostate Cancer
 
Radiotherapy for bladder cancers
Radiotherapy for bladder cancersRadiotherapy for bladder cancers
Radiotherapy for bladder cancers
 
Radiotherapy Planning For Esophageal Cancers
Radiotherapy Planning For Esophageal CancersRadiotherapy Planning For Esophageal Cancers
Radiotherapy Planning For Esophageal Cancers
 
Cytoreductive nephrectomy (when and why)
Cytoreductive nephrectomy (when and why)Cytoreductive nephrectomy (when and why)
Cytoreductive nephrectomy (when and why)
 
Molecular imaging and therapy in prostate cancer
Molecular imaging and therapy in prostate cancerMolecular imaging and therapy in prostate cancer
Molecular imaging and therapy in prostate cancer
 
What Increases Penile Growth Kit
What Increases Penile Growth KitWhat Increases Penile Growth Kit
What Increases Penile Growth Kit
 
Personalised medicine in rt dr. ashutosh
Personalised medicine in rt   dr. ashutoshPersonalised medicine in rt   dr. ashutosh
Personalised medicine in rt dr. ashutosh
 
ECCLU 2011 - S. Supiot - Locally advanced prostate cancer: Multimodality trea...
ECCLU 2011 - S. Supiot - Locally advanced prostate cancer: Multimodality trea...ECCLU 2011 - S. Supiot - Locally advanced prostate cancer: Multimodality trea...
ECCLU 2011 - S. Supiot - Locally advanced prostate cancer: Multimodality trea...
 
Prostate cancer updates 2021
Prostate cancer updates 2021Prostate cancer updates 2021
Prostate cancer updates 2021
 
Radiation therapy in Cancer Oesophagus
Radiation therapy in Cancer Oesophagus Radiation therapy in Cancer Oesophagus
Radiation therapy in Cancer Oesophagus
 
LUNG CANCER MANAGEMENT IN LOW RESOURCE SETTINGS
LUNG CANCER MANAGEMENT IN LOW RESOURCE SETTINGSLUNG CANCER MANAGEMENT IN LOW RESOURCE SETTINGS
LUNG CANCER MANAGEMENT IN LOW RESOURCE SETTINGS
 
ECCLU 2011 - A. Widmark - Prostate cancer: All the truth about local treatmen...
ECCLU 2011 - A. Widmark - Prostate cancer: All the truth about local treatmen...ECCLU 2011 - A. Widmark - Prostate cancer: All the truth about local treatmen...
ECCLU 2011 - A. Widmark - Prostate cancer: All the truth about local treatmen...
 
Oligo final
Oligo finalOligo final
Oligo final
 
Adjuvant radiation based on genomic risk factors emerging scenarios
Adjuvant radiation based on genomic risk factors   emerging scenariosAdjuvant radiation based on genomic risk factors   emerging scenarios
Adjuvant radiation based on genomic risk factors emerging scenarios
 
ROLE OF RADIATION IN BONE TUMORS FOR ORTHOPEDICS
ROLE OF RADIATION IN BONE TUMORS FOR ORTHOPEDICSROLE OF RADIATION IN BONE TUMORS FOR ORTHOPEDICS
ROLE OF RADIATION IN BONE TUMORS FOR ORTHOPEDICS
 
Results of Stereotactic Body Radiotherapy (SBRT) for Management of Hepatic Tu...
Results of Stereotactic Body Radiotherapy (SBRT) for Management of Hepatic Tu...Results of Stereotactic Body Radiotherapy (SBRT) for Management of Hepatic Tu...
Results of Stereotactic Body Radiotherapy (SBRT) for Management of Hepatic Tu...
 
ECCLU 2011 - M. Bolla - Prostate cancer: Locally advanced disease and patient...
ECCLU 2011 - M. Bolla - Prostate cancer: Locally advanced disease and patient...ECCLU 2011 - M. Bolla - Prostate cancer: Locally advanced disease and patient...
ECCLU 2011 - M. Bolla - Prostate cancer: Locally advanced disease and patient...
 
SBRT in head and neck cancer
SBRT in  head and neck cancerSBRT in  head and neck cancer
SBRT in head and neck cancer
 

Viewers also liked

Trabajo descubrimiento ariana
Trabajo descubrimiento arianaTrabajo descubrimiento ariana
Trabajo descubrimiento arianavaleria caicedo
 
Tarea 1 santana javier 14104312
Tarea 1 santana javier 14104312Tarea 1 santana javier 14104312
Tarea 1 santana javier 14104312JAVIER SANTANA
 
08 identity cards to members - 1
08   identity cards to members - 108   identity cards to members - 1
08 identity cards to members - 1spandane
 
арт гимн с,з
арт гимн с,зарт гимн с,з
арт гимн с,зolga080974
 
Practicando musica
Practicando musicaPracticando musica
Practicando musicagisell48
 
Agua contaminada en venezuela
Agua contaminada en venezuelaAgua contaminada en venezuela
Agua contaminada en venezuelajuan_023
 
Julie Foster_HSBC_MP recommendation
Julie Foster_HSBC_MP recommendationJulie Foster_HSBC_MP recommendation
Julie Foster_HSBC_MP recommendationJULIE ERRICKSON
 
Strategi pembelajaran inkuiri dan ekspositori (new)
Strategi pembelajaran inkuiri dan ekspositori (new)Strategi pembelajaran inkuiri dan ekspositori (new)
Strategi pembelajaran inkuiri dan ekspositori (new)Andi Rahim
 
Présentation Mission Terrain 01_08_016_Fr
Présentation Mission Terrain 01_08_016_FrPrésentation Mission Terrain 01_08_016_Fr
Présentation Mission Terrain 01_08_016_FrGondo Gbanyangbe
 
Panorama sites-web-tablettes-iafactory
Panorama sites-web-tablettes-iafactoryPanorama sites-web-tablettes-iafactory
Panorama sites-web-tablettes-iafactoryiafactory
 
Strategi pembelajaran inkuiri
Strategi pembelajaran inkuiriStrategi pembelajaran inkuiri
Strategi pembelajaran inkuiriDiana Dhieant
 
How to deploy KuberDock hassle-free
How to deploy KuberDock hassle-freeHow to deploy KuberDock hassle-free
How to deploy KuberDock hassle-freeCloudLinux
 
Entreprises et Médias Sociaux - Les mardis de la formation AB consultants - f...
Entreprises et Médias Sociaux - Les mardis de la formation AB consultants - f...Entreprises et Médias Sociaux - Les mardis de la formation AB consultants - f...
Entreprises et Médias Sociaux - Les mardis de la formation AB consultants - f...Remy EXELMANS
 

Viewers also liked (16)

Trabajo descubrimiento ariana
Trabajo descubrimiento arianaTrabajo descubrimiento ariana
Trabajo descubrimiento ariana
 
Tarea 1 santana javier 14104312
Tarea 1 santana javier 14104312Tarea 1 santana javier 14104312
Tarea 1 santana javier 14104312
 
08 identity cards to members - 1
08   identity cards to members - 108   identity cards to members - 1
08 identity cards to members - 1
 
арт гимн с,з
арт гимн с,зарт гимн с,з
арт гимн с,з
 
Practica 9 Reloj
Practica 9 RelojPractica 9 Reloj
Practica 9 Reloj
 
Practicando musica
Practicando musicaPracticando musica
Practicando musica
 
Yahoo Brasil
Yahoo BrasilYahoo Brasil
Yahoo Brasil
 
Agua contaminada en venezuela
Agua contaminada en venezuelaAgua contaminada en venezuela
Agua contaminada en venezuela
 
20150731142943
2015073114294320150731142943
20150731142943
 
Julie Foster_HSBC_MP recommendation
Julie Foster_HSBC_MP recommendationJulie Foster_HSBC_MP recommendation
Julie Foster_HSBC_MP recommendation
 
Strategi pembelajaran inkuiri dan ekspositori (new)
Strategi pembelajaran inkuiri dan ekspositori (new)Strategi pembelajaran inkuiri dan ekspositori (new)
Strategi pembelajaran inkuiri dan ekspositori (new)
 
Présentation Mission Terrain 01_08_016_Fr
Présentation Mission Terrain 01_08_016_FrPrésentation Mission Terrain 01_08_016_Fr
Présentation Mission Terrain 01_08_016_Fr
 
Panorama sites-web-tablettes-iafactory
Panorama sites-web-tablettes-iafactoryPanorama sites-web-tablettes-iafactory
Panorama sites-web-tablettes-iafactory
 
Strategi pembelajaran inkuiri
Strategi pembelajaran inkuiriStrategi pembelajaran inkuiri
Strategi pembelajaran inkuiri
 
How to deploy KuberDock hassle-free
How to deploy KuberDock hassle-freeHow to deploy KuberDock hassle-free
How to deploy KuberDock hassle-free
 
Entreprises et Médias Sociaux - Les mardis de la formation AB consultants - f...
Entreprises et Médias Sociaux - Les mardis de la formation AB consultants - f...Entreprises et Médias Sociaux - Les mardis de la formation AB consultants - f...
Entreprises et Médias Sociaux - Les mardis de la formation AB consultants - f...
 

Similar to Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters

Re-irradiation, Prostate Cancer
Re-irradiation, Prostate CancerRe-irradiation, Prostate Cancer
Re-irradiation, Prostate CancerMax Peters
 
Focal Ca prostate.pdf
Focal Ca prostate.pdfFocal Ca prostate.pdf
Focal Ca prostate.pdfssusere131b1
 
Proefschrift Shapiro_defIII_lowres
Proefschrift Shapiro_defIII_lowresProefschrift Shapiro_defIII_lowres
Proefschrift Shapiro_defIII_lowresshapirox
 
Long Term Survival RF Ablation for Primary and Metastatic Liver Tumors
Long Term Survival RF Ablation for Primary and Metastatic Liver TumorsLong Term Survival RF Ablation for Primary and Metastatic Liver Tumors
Long Term Survival RF Ablation for Primary and Metastatic Liver TumorsISWANTO SUCANDY, M.D, F.A.C.S
 
Long term survival radiofrequency ablation for primary and metastatic liver t...
Long term survival radiofrequency ablation for primary and metastatic liver t...Long term survival radiofrequency ablation for primary and metastatic liver t...
Long term survival radiofrequency ablation for primary and metastatic liver t...ISWANTO SUCANDY, M.D, F.A.C.S
 
Discuss the pathology of bladder cancers
Discuss the pathology of bladder cancersDiscuss the pathology of bladder cancers
Discuss the pathology of bladder cancersJim Badmus
 
Metastatic liver disease (2)
Metastatic liver disease (2)Metastatic liver disease (2)
Metastatic liver disease (2)mostafa hegazy
 
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...daranisaha
 
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...JohnJulie1
 
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...eshaasini
 
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...semualkaira
 
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...NainaAnon
 
Clinics of Oncology | Oncology Journals | Open Access Journal
Clinics of Oncology | Oncology Journals | Open Access JournalClinics of Oncology | Oncology Journals | Open Access Journal
Clinics of Oncology | Oncology Journals | Open Access JournalEditorSara
 
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...semualkaira
 

Similar to Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters (20)

Re-irradiation, Prostate Cancer
Re-irradiation, Prostate CancerRe-irradiation, Prostate Cancer
Re-irradiation, Prostate Cancer
 
Focal Ca prostate.pdf
Focal Ca prostate.pdfFocal Ca prostate.pdf
Focal Ca prostate.pdf
 
Proefschrift Shapiro_defIII_lowres
Proefschrift Shapiro_defIII_lowresProefschrift Shapiro_defIII_lowres
Proefschrift Shapiro_defIII_lowres
 
MCC 2011 - Slide 26
MCC 2011 - Slide 26MCC 2011 - Slide 26
MCC 2011 - Slide 26
 
Urology 5th year, 2nd lecture (Dr. Sarwar)
Urology 5th year, 2nd lecture (Dr. Sarwar)Urology 5th year, 2nd lecture (Dr. Sarwar)
Urology 5th year, 2nd lecture (Dr. Sarwar)
 
Long Term Survival RF Ablation for Primary and Metastatic Liver Tumors
Long Term Survival RF Ablation for Primary and Metastatic Liver TumorsLong Term Survival RF Ablation for Primary and Metastatic Liver Tumors
Long Term Survival RF Ablation for Primary and Metastatic Liver Tumors
 
Long term survival radiofrequency ablation for primary and metastatic liver t...
Long term survival radiofrequency ablation for primary and metastatic liver t...Long term survival radiofrequency ablation for primary and metastatic liver t...
Long term survival radiofrequency ablation for primary and metastatic liver t...
 
Discuss the pathology of bladder cancers
Discuss the pathology of bladder cancersDiscuss the pathology of bladder cancers
Discuss the pathology of bladder cancers
 
Metastatic liver disease (2)
Metastatic liver disease (2)Metastatic liver disease (2)
Metastatic liver disease (2)
 
IJET-V3I2P22
IJET-V3I2P22IJET-V3I2P22
IJET-V3I2P22
 
Portec 3
Portec 3Portec 3
Portec 3
 
How to Integrate Perioperative Immunotherapy Into Multimodal Treatment Plans ...
How to Integrate Perioperative Immunotherapy Into Multimodal Treatment Plans ...How to Integrate Perioperative Immunotherapy Into Multimodal Treatment Plans ...
How to Integrate Perioperative Immunotherapy Into Multimodal Treatment Plans ...
 
Oesophagus Cancer.
Oesophagus Cancer.Oesophagus Cancer.
Oesophagus Cancer.
 
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
 
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
 
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
 
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
 
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
 
Clinics of Oncology | Oncology Journals | Open Access Journal
Clinics of Oncology | Oncology Journals | Open Access JournalClinics of Oncology | Oncology Journals | Open Access Journal
Clinics of Oncology | Oncology Journals | Open Access Journal
 
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
Upper Rectal Cancer: Benefit After Preoperative Chemoradiation Versus Upfront...
 

More from Max Peters

Who can do without patients
Who can do without patientsWho can do without patients
Who can do without patientsMax Peters
 
Urethral and bladder dosimetry of total and focal salvage Iodine-125 prostate...
Urethral and bladder dosimetry of total and focal salvage Iodine-125 prostate...Urethral and bladder dosimetry of total and focal salvage Iodine-125 prostate...
Urethral and bladder dosimetry of total and focal salvage Iodine-125 prostate...Max Peters
 
Rectal dose constraints for salvage iodine-125 prostate brachytherapy.
Rectal dose constraints for salvage iodine-125 prostate brachytherapy.Rectal dose constraints for salvage iodine-125 prostate brachytherapy.
Rectal dose constraints for salvage iodine-125 prostate brachytherapy.Max Peters
 
Multivariable model development and internal validation for prostate cancer s...
Multivariable model development and internal validation for prostate cancer s...Multivariable model development and internal validation for prostate cancer s...
Multivariable model development and internal validation for prostate cancer s...Max Peters
 
Development and internal validation of a multivariable prediction model for b...
Development and internal validation of a multivariable prediction model for b...Development and internal validation of a multivariable prediction model for b...
Development and internal validation of a multivariable prediction model for b...Max Peters
 
Bedside teaching in medical education; a literature review.
Bedside teaching in medical education; a literature review.Bedside teaching in medical education; a literature review.
Bedside teaching in medical education; a literature review.Max Peters
 

More from Max Peters (6)

Who can do without patients
Who can do without patientsWho can do without patients
Who can do without patients
 
Urethral and bladder dosimetry of total and focal salvage Iodine-125 prostate...
Urethral and bladder dosimetry of total and focal salvage Iodine-125 prostate...Urethral and bladder dosimetry of total and focal salvage Iodine-125 prostate...
Urethral and bladder dosimetry of total and focal salvage Iodine-125 prostate...
 
Rectal dose constraints for salvage iodine-125 prostate brachytherapy.
Rectal dose constraints for salvage iodine-125 prostate brachytherapy.Rectal dose constraints for salvage iodine-125 prostate brachytherapy.
Rectal dose constraints for salvage iodine-125 prostate brachytherapy.
 
Multivariable model development and internal validation for prostate cancer s...
Multivariable model development and internal validation for prostate cancer s...Multivariable model development and internal validation for prostate cancer s...
Multivariable model development and internal validation for prostate cancer s...
 
Development and internal validation of a multivariable prediction model for b...
Development and internal validation of a multivariable prediction model for b...Development and internal validation of a multivariable prediction model for b...
Development and internal validation of a multivariable prediction model for b...
 
Bedside teaching in medical education; a literature review.
Bedside teaching in medical education; a literature review.Bedside teaching in medical education; a literature review.
Bedside teaching in medical education; a literature review.
 

Digital version thesis Salvage for radiorecurrent prostate cancer, Max Peters

  • 1. Salvageforradiorecurrentprostatecancer MaxPeters2016 Salvage for radiorecurrent prostate cancer Max Peters Patients undergoing primary radiotherapy for prostate cancer are at risk of recurrent disease. Recurrences are often confined to the prostate and therefore eligible for a second curative ablation, known as salvage. Salvage modalities are usually directed at the entire prostate volume, due to former difficulties in assessing localised recurrences. Because of previous radiation damage to the prostate and surrounding organs at risk (OAR), whole-gland salvage modalities are associated with significant toxicity rates. As a result of recent developments in assessing prostate cancer location using multi- parametric MRI and different biopsy techniques, the attention has shifted to a focal treatment approach in the salvage setting. This is known as focal salvage. With this approach only the recurrent tumour area is targeted, which seems to maintain cancer control, but decreases toxicity rates. Focal salvage Iodine-125 brachytherapy appears to be able to provide durable disease control, while minimising late severe genitourinary (GU) and gastrointestinal (GI) toxicity. However, for salvage brachytherapy in general, dose constraints are necessary for OAR to prevent late severe complications. In the primary setting, these are available for both late GU and GI toxicity. These restrictions are probably inaccurate for the salvage setting, since the repair capacity of OAR is compromised by previous radiation damage. The dose restrictions for the rectum, urethra and bladder as found in this thesis are lower than those used for primary brachytherapy. Before implementation of these restrictions is possible, patients should be adequately selected for salvage. Failure rates for whole-gland salvage are often significant, indicating a role for prediction models assessing which patients are most eligible for treatment. Whole-gland salvage brachytherapy patients can be adequately selected using the disease free survival interval (DFSI) after primary therapy and their PSA- doubling time (PSADT) before salvage. Forfocalsalvage,therearenomultivariablemodelsavailablebecauseseriesareusually limited. The model in this thesis for focal salvage high intensity focused ultrasound (HIFU) shows that the DFSI after primary radiotherapy, the pre-treatment PSA, PSADT, prostate volume and T-stage as assessed on MRI can adequately predict biochemical disease free survival up to 3 years follow-up. Lastly, it seems focal salvage brachytherapy is the most cost-effective option, predominantly driven by the lower treatment costs and reduced toxicity rates for which follow-up treatment is indicated.
  • 2.
  • 4. Salvage for radiorecurrent prostate cancer PhD thesis, Utrecht University, the Netherlands Copyright M. Peters, Utrecht, 2016 All rights reserved. No part of this publication may be reproduced, distributed, or transmitted in any form or by any means, including photocopying, recording, or other electronic or mechanical methods, without the prior written permission of the publisher, except in the case of brief quotations embodied in critical reviews and certain other noncommercial uses permitted by copyright law. Copyright of the published or accepted articles has been transferred to the respective journals. The research described in this thesis was performed at the department of Radiation Oncology of the University Medical Centre Utrecht. Cover: Gold of the Azure by Joan Miró (1967), edited by Roy Sanders. Lay out: Roy Sanders Print: Print Support4U ISBN: 978-90-821-9683-2 Copyright: Chapter 1 Springer-Verlag GmbH, Berlin © 2013 Chapter 3 Elsevier Ireland Ltd © 2014 Chapter 4 Elsevier Ireland Ltd. © 2015 Chapter 5 American Brachytherapy Society © 2015 Epub ahead of print Chapter 6 American Brachytherapy Society © 2016 Accepted Chapter 7 Elsevier Ireland Ltd. © 2016 Accepted
  • 5. Salvage for radiorecurrent prostate cancer Herbehandeling van prostaatkanker recidieven na radiotherapie (met een samenvatting in het Nederlands) Proefschrift ter verkrijging van de graad van doctor aan de Universiteit Utrecht op gezag van de rector magnificus, prof.dr. G.J. van der Zwaan, ingevolge het besluit van het college voor promoties in het openbaar te verdedigen op donderdag 10 maart 2016 des middags te 2.30 uur door Max Peters geboren op 17 mei 1989 te Utrecht
  • 6. Promotor: Prof. dr. M. van Vulpen Copromotoren: Dr. J.R.N. van der Voort van Zyp Dr. Ir. M.A. Moerland Dit proefschrift werd (mede) mogelijk gemaakt door financiële steun van Varian Medical Systems, Elekta en de prostaatkankerstichting
  • 7. “History repeats, but science reverberates.” Siddhartha Mukherjee, The Emperor of All Maladies: A Biography of Cancer
  • 8. 33 Chapter 1 Patterns of outcome and toxicity after salvage prostatectomy, salvage cryosurgery and salvage Iodine-125 brachytherapy for prostate cancer recurrences after radiation therapy: a multi-centre experience and literature comparison. Contents 09 Introduction and outline of the thesis 49 Chapter 2 Focal salvage therapy for local prostate cancer recurrences after primary radiotherapy – a comprehensive review. 67 Chapter 3 Focal salvage Iodine-125 brachytherapy for prostate cancer recurrences after primary radiotherapy: a retrospective study regarding toxicity, biochemical outcome and quality of life. 89 Chapter 4 Urethral and bladder dosimetry of total and focal salvage Iodine-125 prostate brachytherapy: Late toxicity and dose constraints. 109 Chapter 5 Rectal dose constraints for salvage Iodine-125 prostate brachytherapy.
  • 9. 127 Chapter 6 Development and internal validation of a multivariable prediction model for biochemical failure after whole-gland salvage Iodine-125 prostate brachytherapy for recurrent prostate cancer. 147 Chapter 7 Multivariable model development and internal validation for prostate cancer specific survival and overall survival after whole-gland salvage Iodine-125 prostate brachytherapy. 167 Chapter 8 Development and internal validation of a multivariable prediction model for biochemical failure after focal salvage high intensity focused ultrasound for locally recurrent prostate cancer: presentation of a risk score for individual patient prognosis. 187 Chapter 9 Comparative cost-effectiveness of focal and total salvage I-125 brachytherapy for recurrent prostate cancer after primary radiotherapy. 203 Chapter 10 Discussion Summary 233 Addenda Samenvatting Dankwoord List of publications Curriculum vitae
  • 10.
  • 12. Introduction and outline 10 1. Epidemiology In many countries, prostate cancer is the most commonly diagnosed male cancer (excluding non-melanoma skin cancers), with an estimated 220,800 new cases and an expected 27,540 prostate cancer related deaths in the United States in 20151 . In the Netherlands, there were approximately 10.000 new cases in 2014. Since the 1990’s, the annual mortality rate is approximately stable at 2500 yearly2 . In 2012, there were 1.1 million cases worldwide, with 307,000 attributable deaths3 . Prostate cancer is often diagnosed in an early stage and these patients are usually eligible for active surveillance or radical treatment. However, because of the high incidence, in absolute terms it remains the cancer site with the second most cancer related deaths in the United States (after lung cancer) and the fifth most common cause of cancer-related death worldwide1,3 . 2. Stage migration As opposed to many other types of cancer, there has been a dramatic rise in the incidence of prostate cancer in the past decades, as can be seen in figure 1. Due to several factors, a significant and increasing proportion of patients is diagnosed in an early and often organ-confined stage. Most importantly, the dramatic rise in incidence of localised disease is a direct consequence of the introduction of PSA- testing in the 1990’s (figure 1)1 . Patients are screened using PSA at an earlier age and more frequently, thereby increasing the possibility of finding indolent, organ- confined tumours. Figure 1: Time-trends in several tumour incidences. Noticeable is the dramatic rise in prostate cancer incidence1   Male Prostate Lung & bronchus Colorectum Urinary bladder Thyroid Liver*Melanoma of the skin Rateper100,000Population 0 25 50 75 100 125 150 175 200 225 250 1980 1985 1990 1995 2000 2005 2011
  • 13. Introduction and outline 11 Localised disease is usually assessed using digital rectal examination (DRE) in combination with transrectal ultrasound (TRUS)-guided biopsies. Technetium-99 scintigraphy (bone-scan) and/or CT are used to exclude metastases in high risk localised disease (with a Gleason score ≥8 and/or PSA ≥20) or ≥cT3a tumours4 . In addition,localiseddiseasecanbepartlypredictedbasedonnomogramsincorporating clinical characteristics: for example Partin tables which use the clinical tumour(T)- stage, histopathological Gleason score and the PSA-value5 . In recent years, however, developments in diagnostic techniques have led to increasing accuracy in assessing localised disease and partly in excluding lymph- node or distant metastases. Multiparametric (mp) magnetic resonance imaging (MRI) and 11C-Choline and 18F-Fluorocholine Positron-Emission Tomography in combinationwithCT(PET-CT)havecontributedsignificantlytothisassessment4,6-14 . Together with developments in biopsy techniques such as transperineal template prostate mapping (TTPM) and MRI-guided biopsies, there has been a further increaseinclassifyinglocalisedandclinicallysignificantdiseaseoverDREandTRUS- guided biopsies11,12,15-18 . This stage migration to organ-confined and increasingly low- risk disease has some important consequences: most importantly the potential to curatively treat disease and thereby excluding or postponing the use of androgen deprivation therapy (ADT) in case of metastases. 3. To treat or not to treat? 3.1 Treatment versus observation Whether these often low-volume, low-grade tumours need radical treatment is a challenging question. Large randomised controlled trials (RCT’s) have provided conflicting results regarding radical treatment for early-stage prostate cancer. The Prostate Cancer Intervention versus Observation (PIVOT)-trial found no evidence of a survival difference in patients with localised prostate cancer who were randomised between radical prostatectomy and watchful waiting (hazard ratio [HR] 0.88; 95% confidence interval [CI] 0.71-1.08; p=0.22; absolute risk reduction 2.9%19 ). Contrary, the Scandinavian Prostate Cancer Group Study Number 4 (SPCG-4) did find a difference in both overall and prostate cancer specific mortality, with a number needed to treat (NNT) of approximately 8 to prevent one death20 . However, the PIVOT-study might have been exposed to the above mentioned stage migration, since patients were recruited mostly in the PSA-era. This was primarily visible in the incidence of 50% non-palpable T1c-tumours. The SPGC-4 study only had 12% T1c tumours, because patients were assessed mostly before the PSA era. This could suggest a possible delayed effect of radical prostatectomy in the PIVOT trial which might become visible after longer follow-up.
  • 14. Introduction and outline 12 3.2 Screening This so-called ‘lead-time bias’ also seems to be present in large screening trials for prostate cancer, in which the ‘number needed to screen (NNS)’ and ‘number needed to treat (NNT)’ to prevent one death due to prostate cancer seem to be decreasing with longer follow-up. This has been observed in the European Randomised Study of Screening for Prostate Cancer (ERSPC), in which over the years the NNS and NNT have decreased from 1410 and 48 to 781 and 27, respectively21-23 . However, not all studies confirm these results, with the most important example being the US Prostate, Lung, Colorectal, and Ovarian (PLCO) cancer screening trial, which showed no effect of annual PSA and DRE on survival. This study, however, suffered from a significant contamination of the control group, in which 50% of the patients underwent PSA-testing outside the study protocol24 . A recent Cochrane meta-analysis did not show a pooled effect of PSA-screening in five studies (including the ERSPC and PLCO) on prostate cancer specific and overall mortality25,26 . This effect was also not present when pooling the results of the ERSPC and PLCO separately. In addition, no effect on overall mortality has been observed in any of the separate trials. 3.3 Side-effects of treatment Assuming an effect of screening, the question remains whether the latest NNS of 781 and NNT of 27 from the ERSPC are worth the often severe side-effects of primary radical treatment. Toxicity data from the PIVOT trial shows significant incontinence rates after treatment: 17.1% in the radical prostatectomy group versus 6.3% in the observation group. Erectile dysfunction also considerably increased: 81.1% versus 44.1%. In addition, over 20% often severe 30-day surgical morbidity was seen. Aggravation of toxicity after radical treatment is seen up to 15 years, as assessed in a large cohort study comparing radiotherapy with prostatectomy (n=1655). Both prostatectomy and radiotherapy showed an incontinence rate of approximately 18% in this cohort and 90% erectile dysfunction. Radiotherapy patients had some more bowel dysfunction at 15 years follow-up (16% versus 5.2%)27 . Whether these effects are actually related to surgery or radiotherapy will possibly be answered by the awaited results of the ProtecT trial, an RCT evaluating radiotherapy, radical prostatectomy and active surveillance for localised disease. In addition, this trial is the first comparing these treatment modalities in a direct manner. Active surveillance in the current era seems a safe option for low risk patients, with long term excellent prostate cancer specific survival while preventing side-effects from radical treatment28 . Especially with the increasing use of PSA as
  • 15. Introduction and outline 13 a screening tool and therefore a large stage migration to low-risk disease, active surveillance is appealing both in terms of quality of life as well as cost-effectiveness. Whether treatment is necessary for all patients therefore remains a vital question. 4. Radiotherapy as primary treatment When we choose to treat, there is little evidence from randomised controlled trials that suggests how to treat. As the leading modalities for primary treatment, radical prostatectomy and radiotherapy do not differ substantially in terms of cancer control29 . This would make the side-effect profile essential, which might favour radiotherapy. There are several developments in the field of radiotherapy that make the treatment appealing. Using external beam radiotherapy (EBRT), intensity modulated radiotherapy (IMRT) offers a great advantage over the older 3D conformal technique (3D-CRT) by increasing the dose delivered to the prostate while sparing organs at risk (OAR)30,31 . The use of IMRT for treatment of patients with localised prostate cancer has therefore increased from 0.15% in 2000 to 95.6% in 200832 . A recent development is that proton therapy is increasingly being adopted, because of its theoretical advantages in increasing conformality. All therapies show high biochemical disease free survival (bDFS)29 . However, preliminary results seem to favour IMRT over proton therapy in terms of treatment-related side-effects, especially gastrointestinal32,33 . Furthermore, combining EBRT with brachytherapy, it is possible to boost the tumour,whichisadvantageousespeciallyforintermediateandhighriskpatients34,35 . These improvements in accuracy of dose delivery resulted in over 40% of prostate cancer patients older than 65 year nowadays being treated with EBRT, brachytherapy or a combination36 . Where EBRT is traditionally delivered fractionated, brachytherapy can be delivered in a single procedure. In low and intermediate risk patients, low-dose- rate (LDR) and high-dose-rate (HDR) brachytherapy can achieve bDFS rates that are comparable to other primary treatment modalities29,35,37 . Brachytherapy offers a steep dose decline and high conformality, limiting damage to surrounding organs at risk and subsequently preventing toxicity. Also the single procedure in which it can be performed has a great benefit over the many daily fractions with external beam radiotherapy38 .This makes brachytherapy the most cost-effective primary treatmentmodalityforprostatecancer39 .Arecentpropensityscoreadjustedanalysis even indicated that brachytherapy leads to significantly increased bDFS compared to EBRT for low and intermediate risk patients40 . Unfortunately, brachytherapy as
  • 16. Introduction and outline 14 a primary treatment modality seems to be declining due to increasing popularity of other primary (more expensive) treatments, among other factors38,41 . Another advantage of brachytherapy that was developed in recent years, is a focal approach to prostate cancer, both with LDR42 and HDR (Netherlands Trial register number3790andNCT00913939),asopposedtotraditionalwhole-glandtreatment. This was made possible by increased accuracy of tumour localisation using multiparametric MRI and (image-guided or systematic) biopsy techniques8,10,16,43,44 . Also applied with modalities like high intensity focused ultrasound (HIFU)45,46 and cryotherapy47,48 , the focal approach appears to substantially decrease toxicity rates while possibly maintaining acceptable cancer control49 . However, longer follow- up and randomised trials are necessary to ascertain this, because proponents and opponents considerably differ in their viewpoints49-51 . Due to this large range of developments in primary radiotherapy and their effects on prostate cancer management, radiotherapy is increasing as a primary treatment modality. 5. Recurrent disease after primary radiotherapy While whole-gland radiotherapy is an established and effective treatment option for primary tumours, recurrences can still be common. Even in the era of dose escalation, in which total treatment doses are above 78 Gray (Gy) in the majority of patients, biochemical relapses occur frequently52-54 . The Kaplan-Meier curves in figure 2 show the biochemical recurrence rates for the three National Cancer Comprehensive Network (NCCN) risk groups. Estimated 8-year recurrence rates of 9.7%, 22.7% and 43.9% for low, intermediate and high risk groups were observed, respectively. All 2.694 patients were treated with IMRT with a total dose of ≥79.2 Gy, up to maximal 85.6 Gy53 . The development of biochemical failure is problematic, since it commonly precedes the development of distant metastases by a median 5.4 years and prostate cancer specific mortality by a median 10.5 years53 . Even though cancer control rates are increasing with further dose-escalation, ADT-use and enhanced patient selection, there will be an increase in absolute numbers of patients with biochemical recurrent disease because more patients are primarily treated with radiotherapy for prostate cancer. In addition, the previously discussed rise in PSA-screening will shift the diagnosis of prostate cancer to an increasingly lower age, creating a group with a longer life-expectancy after primary treatment and therefore possibly more risk of developing biochemical failure. Basically, physicians will more often be faced with biochemical recurrent disease in the future and management of these recurrences will become an important issue.
  • 17. Introduction and outline 15 6. The index lesion and the relation with recurrent disease Biochemical recurrence in prostate cancer often seems to be the result of a localised process. The data on the exact location of these recurrences is scarce, but in several series it is suggested that the tumour often regrows at the site of the primary dominant and usually largest lesion, also called the ‘index lesion’ (figure 3)55-59 .The evidence is not straightforward, however, with some suggesting tumour regrowth to occur more multifocal, although organ-confined and even unifocal disease is still often observed60,61 . Furthermore, salvage radical prostatectomy is not always used as the reference standard to assess the localised radiorecurrent tumours, thereby possibly not giving a representable estimate of the exact recurrence location(s). In addition, patients were often treated with lower doses of radiation than with current dose-escalation schedules. This could mean that radiorecurrent disease is currently shifting towards the index lesion because secondary tumour foci are increasingly successfully treated55,57,58 . This idea of the index lesion has originated in the primary setting, where it represents the tumour driving the prognosis of the disease. While prostate cancer is often a multifocal disease with multiple, often bilateral foci62 , the index lesion is thought in most cases to make up the largest and most aggressive tumour63-65 . Figure 2: Biochemical recurrence rates per NCCN risk group53 Low risk 590 547 471 363 263 129 58 19 Int Risk 1289 1129 880 566 354 200 86 24 High Risk 815 653 488 337 226 133 66 29 Low risk Intermediate Risk High Risk PSARecurrenceFreeSurvival Time (yr) 100% 80% 60% 40% 20% 0% 0 2 4 6 8 10 12 14
  • 18. Introduction and outline 16 It has even been observed that prostate cancer metastases have a monoclonal singular origin which could originate in the index lesion, although this is still being (fiercely) debated66,67 . This idea of the index lesion is tempting, since targeting therapy to only a focal region of the prostate might reduce toxicity associated with whole-gland treatment45,46,49,68-71 . As discussed above, treatments in the primary setting are increasingly targeting partial areas of the gland72,73 , or boosting dominant intraprostatic lesions74 . Trials are even ongoing in which ‘insignificant’ tumour foci are left untreated45 . Significant developments in staging procedures using multiparametric-MRI and sophisticated biopsy-techniques make such an alteration in the way we think of and treat prostate cancer possible. Proponents of the focal approach compare it to other tumour sites in which this is now common practice, such as breast and kidney cancer75 . It has even been called a ‘male lumpectomy’ in the cryotherapy setting48 , equivalent to breast cancer treatment. For recurrent tumours, this idea of the index lesion and a focal treatment approach is potentially even more appealing, since secondary tumour foci might have been successfully treated by the primary radiation. In addition, the decrease in treatment-related toxicity is potentially much larger in the recurrent treatment setting. These ideas are gaining increasing international attention and will be further explored here. Figure 3: Recurrent prostate tumours. Depicted here as vertical lines (over the horizontal lines indicating the primary lesion)55  
  • 19. Introduction and outline 17 7. Current treatment strategies for recurrent disease This thesis will focus on the specific subgroup of patients who have locally failed primary radiotherapy. In prostate cancer, treating localised radiorecurrent prostate cancer with a curative second approach is referred to as salvage. There are quite extensive series available in the literature, describing a variety of salvage procedures. Mostly, these procedures are directed at the entire prostatic volume. The largest series available to date are those of salvage radical prostatectomy (SRP)76-78 , salvage cryosurgery (SCS)79-81 , salvage HIFU82,83 and salvage brachytherapy (SBT, both LDR and HDR)84-87 . All series show decent cancer control rates, with 5-year bDFS in the range of 50-60% for SRP, SCS and SBT and 40-50% after 3 years for salvage HIFU88-91 . These techniques therefore have the potential to postpone, or even prevent the use of ADT, which has been associated with significant cardiovascular, endocrinologic and sexual toxicity and even mental decline92,93 . However, a recent overview has shown that salvage therapies are not frequently adopted, with approximately 98% of patients receiving ADT as subsequent treatment for recurrent disease. Only 2% of patients were found to be treated with a form of curatively intended salvage94 . This was also observed in an analysis of the large (UCSF) Cancer of the Prostate Strategic Urological Research Endeavor (CaPSURE) database95 . A reason to refrain from salvage therapies is that they are mostly performed on a whole-gland basis88,90,91,96 . Because of prior difficulties in the assessment of recurrences, whole gland approaches were (and are still often) the only way to make sure residual disease is fully targeted. The downside to a whole-gland approach is the severe toxicity associated with these procedures. After primary radiotherapy, the prostate becomes fibrotic and OAR surrounding the prostate such as the rectal wall, bladder neck and urethra, show similar changes85,97 . These fibrotic changes impair the normal repair capacities of these structures, making salvage surgical approaches difficult and cumulative radiation from SBT hazardous. High percentages of toxicity have been found with all salvage modalities, although with a wide spread across series. Summarised, up to a third of patients can experience a combination of severe (≥grade 3) genitourinary (GU) and gastro-intestinal (GI) toxicity, requiring surgical intervention to resolve89,91,96,98 . Erectile dysfunction is poorly reported and frequently pre-existent, but often found to be almost universal after salvage procedures88,96,98 . These severe side-effects are likely to contribute to the previously mentioned 2% of patients who are treated with some form of salvage. Salvage has for these reasons remained a treatment generally offered only in specialised, high volume centres.
  • 20. Introduction and outline 18 8. Focal salvage Due to the severe toxicity rates associated with whole-gland salvage in combination with the developments in accurate staging and the attention on focally targeting prostate cancer, the hypothesis of a focal approach in the salvage setting is being developed. With the evidence that recurrences are often localised at the primary index lesion and secondary tumour foci are successfully treated by the primary radiation, the rationale for a focal approach might be even more potent in the salvage than in the primary setting. A focal approach could theoretically spare a substantial part of the prostate and with this, prevent damage to surrounding OAR. However, this procedure is technically challenging and the work-up to assess focal recurrences is intensive and requires a high level of specialised assessment99 . Therefore, focal salvage series are limited in the literature and performed only in specialised centres. A variety of techniques are now described in some pilot series, most importantly cryotherapy (n=10-91)100-103 , HIFU (n=39-48)104,105 and Iodine-125 (LDR) brachytherapy (n=15-25)106-108 . Most of these series adopt a multimodality approach in assessing recurrent disease. This consists of multiparametric MRI: T2-weighted, dynamic contrast enhanced (DCE), diffusion weighted imaging (DWI) and sometimes spectroscopy (MRSI). Furthermore, tumour location is confirmed by either systematic transrectal ultrasound guided biopsies, or template prostate mapping biopsies (TTPM) in one series104 . The extent of ablation varies from completely focal with a specified margin to hemi or zonal, which is also referred to as a ‘dog leg’ or ‘hockey stick’ ablation (Figure 4)73 . Results from focal salvage series are preliminary, but seem favourable. Reported focal salvage 1, 2, 3 and 5- year bDFS rates are 69%-100%, 49%-100%, 50%-72.4% and 46.5%-54.4%, respectively101-108 . These are in line with bDFS-outcomes from   Targeted ablation Hemiablation Zonal ablation Figure 4: Different focal ablation methods for the primary and salvage setting73 .
  • 21. Introduction and outline 19 the whole-gland setting. However, a selection bias towards a more favourable focal salvage patient group cannot be excluded based on these observational studies. Randomised trials, or (less favourably) propensity-score adjusted observational cohorts will have to assess whether there are differences in cancer control between a whole-gland and focal salvage approach. Nonetheless, even if cancer control rates are in favour of whole-gland salvage, the decrease in toxicity with focal salvage could still be a reason to refrain from a whole- gland approach. Toxicity results from focal salvage are favourable, with most studies reporting side-effects which are self-limiting or treatable with medication (grade 1 and 2)101,103,106,107 . Some series report higher toxicity rates, which sometimes need surgical intervention to resolve104,108 . The extent of treatment could have influenced these results, with one study targeting the entire peripheral zone108 . If toxicity results are favourable compared to whole-gland approaches, repeating focal salvage could even be an option if patients initially fail retreatment. This has been successfully done in one of the focal SBT series106 and focal salvage cryotherapy series100 . 9. Problems with current whole-gland and focal salvage I-125 brachytherapy Brachytherapy is often adopted as a salvage modality, both in the whole-gland and focal savage setting. To perform SBT in a safe and effective way, dose constraints and selection criteria related to cancer control are of vital importance. However, these are not available in the current literature. Dose constraints are usually adopted from the primary setting. However, these restrictions might be too lenient because of previous radiation damage to surrounding organs at risk109-113 . It is vital that there are constraints found for SBT below which toxicity is reduced to a minimum, in a way equivalent to the research which is available for primary brachytherapy111,112,114 . Selection criteria in relation to cancer control are available from multivariable analyses performed in larger whole-gland salvage cohorts using SRP, SCS and HIFU76,79,83 . For whole-gland SBT, these are less well defined, due to smaller series with limited events to which prognostic determinants can be related85-87,115 . Also, predictive factors are often categorised (leading to information loss), missing data is not described and no prediction models are available to obtain adequate individual prognoses. These problems are also observed in the other salvage modalities. Furthermore, due to the limited focal salvage data so far, there has not been any uni- or multivariable modelling done to identify factors related to cancer control. For these reasons, selection of patients for whole-gland and especially focal
  • 22. Introduction and outline 20 salvage is now often performed on the basis of determinants related to prognosis in the primary setting or on expert opinion. Conclusively, to this day there are no reliable dose constraints, selection criteria and prediction models developed specifically for SBT. They are frequently adopted from primary BT data and do not take into account issues such as previous radiation damage to OAR or the clinical profile of patients. 10. Outline This thesis will focus on cancer control and toxicity regarding whole-gland salvage modalities and focal salvage Iodine-125 brachytherapy and HIFU. Chapter 1 demonstrates that whole-gland salvage modalities adopted in the Netherlands nowadays are associated with high failure rates, probably because of insufficient assessment of localised disease and exclusion of metastases. Also, significant toxicity rates are observed, which is probably due to targeting the entire prostatic volume. To prevent the high toxicity rates of these whole-gland approaches while maintaining cancer control, focal salvage might be a viable solution. The literature provides us with small series assessing different focal salvage modalities. Chapter 2 summarises literature results of focal and whole- gland salvage and compares cancer control and toxicity. A focal salvage approach using I-125 (LDR) brachytherapy has further been evaluated at our department. Twenty patients with a biochemical recurrence after primary radiotherapy (EBRT or I-125 brachytherapy) were treated. Cancer control, toxicity and quality of life of this cohort are evaluated in chapter 3. The whole-gland I-125 brachytherapy cohort from chapter 1 and the focal salvage cohort from chapter 3 were further evaluated regarding dosimetry and toxicity of both the genitourinary (GU) and gastro-intestinal (GI) system. With these dosimetry differences it was possible to construct dose constraints to prevent late severe GU and GI toxicity (chapters 4 and 5, respectively). Dose constraints for salvage I-125 brachytherapy are important for future adoption of this technique, since constraints for the primary setting might be too lenient in salvage patients because of previous radiation damage to surrounding organs at risk. Chapters 6, 7 and 8 are all related to prediction models to improve selection of patients for both whole-gland and focal salvage. Selection is often performed based on characteristics associated with failure in the primary setting. However, whether these characteristics have the same predictive value in the salvage setting is mostly unknown. No prediction models to quantify the exact effects of pre-treatment characteristics are available for salvage. For the larger whole-gland salvage series,
  • 23. Introduction and outline 21 there are some multivariable models assessing the effects of certain characteristics. For SBT, these models are based on small datasets with often limited events, categorisation of predictors, insufficient description of missing data and other problems which lead to an inaccurate assessment of the effect of prognostic factors. To overcome these problems, chapters 6 and 7 use the most recent methodological guidelines to quantify the effect of prognostic factors on both biochemical failure (chapter 6) and mortality (chapter 7, both prostate cancer specific and overall mortality). With these prediction models, a more accurate selection of whole- gland SBT patients is possible. Nevertheless, these predictors are assessed for whole-gland SBT and might have a different relation with the outcome in focal salvage. The focal SBT cohort from chapter 3 was too limited to create a prediction model. Therefore, in cooperation with the University College London Hospitals (UCLH) Department of Urology, a prediction model was created based on their focal salvage HIFU cohort after EBRT-failure. This prediction model was based on a substantially larger cohort and therefore the creation of a clinically useful risk score was possible. These results are described in chapter 8. Lastly, if focal salvage leads to equivalent cancer control rates and less toxicity than whole-gland salvage, cost-effectiveness is an important aspect which needs to be assessed. Chapter 9 compares the results of the whole-gland and focal salvage I-125 brachytherapy cohorts in terms of cost-effectiveness. The limitations of the described research and future perspectives with regard to salvage are discussed after these chapters in the general discussion.
  • 24. Introduction and outline 22 References 1. Siegel RL, Sahar L, Portier KM, Ward EM, Jemal A. Cancer death rates in US congressional districts. CA Cancer J Clin. 2015. doi: 10.3322/caac.21292 [doi]. 2. Integraal kankercentrum Nederland. Incidentie prostaatkanker. http://www.cijfersoverkanker. nl/selecties/Incidentie_prostaatkanker/img54d0ecdf1c883. Updated 2014. Accessed 09/25, 2015. 3. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359-86. doi: 10.1002/ijc.29210 [doi]. 4. Integraal Kankercentrum Nederland. Richtlijn prostaatcarcinoom. http://www.oncoline.nl/ richtlijn/doc/index.php?type=pda&richtlijn_id=934. Updated 2014. Accessed 09/01, 2015. 5. Partin AW, Mangold LA, Lamm DM, Walsh PC, Epstein JI, Pearson JD. Contemporary update of prostate cancer staging nomograms (partin tables) for the new millennium. Urology. 2001;58(6):843-848. doi: S0090429501014418 [pii]. 6. Barentsz JO, Richenberg J, Clements R, et al. ESUR prostate MR guidelines 2012. Eur Radiol. 2012;22(4):746-757. doi: 10.1007/s00330-011-2377-y [doi]. 7. Umbehr MH, Muntener M, Hany T, Sulser T, Bachmann LM. The role of 11C-choline and 18F-fluorocholine positron emission tomography (PET) and PET/CT in prostate cancer: A systematic review and meta-analysis. Eur Urol. 2013;64(1):106-117. doi: 10.1016/j. eururo.2013.04.019 [doi]. 8. Futterer JJ, Briganti A, De Visschere P, et al. Can clinically significant prostate cancer be detected with multiparametric magnetic resonance imaging? A systematic review of the literature. Eur Urol. 2015. doi: S0302-2838(15)00036-6 [pii]. 9. Hamoen EH, de Rooij M, Witjes JA, Barentsz JO, Rovers MM. Use of the prostate imaging reporting and data system (PI-RADS) for prostate cancer detection with multiparametric magnetic resonance imaging: A diagnostic meta-analysis. Eur Urol. 2015;67(6):1112-1121. doi: 10.1016/j.eururo.2014.10.033 [doi]. 10. de Rooij M, Hamoen EH, Witjes JA, Barentsz JO, Rovers MM. Accuracy of magnetic resonance imaging for local staging of prostate cancer: A diagnostic meta-analysis. Eur Urol. 2015. doi: S0302-2838(15)00674-0 [pii]. 11. Onur R, Littrup PJ, Pontes JE, Bianco FJ,Jr. Contemporary impact of transrectal ultrasound lesions for prostate cancer detection. J Urol. 2004;172(2):512-514. doi: S0022-5347(05)61673-4 [pii]. 12. Schroder FH, van der Maas P, Beemsterboer P, et al. Evaluation of the digital rectal examination as a screening test for prostate cancer. rotterdam section of the european randomized study of screening for prostate cancer. J Natl Cancer Inst. 1998;90(23):1817-1823. 13. Abuzallouf S, Dayes I, Lukka H. Baseline staging of newly diagnosed prostate cancer: A summary of the literature. J Urol. 2004;171(6 Pt 1):2122-2127. doi: 00005392-200406000-00006 [pii]. 14. Evangelista L, Zattoni F, Guttilla A, et al. Choline PET or PET/CT and biochemical relapse of prostate cancer: A systematic review and meta-analysis. Clin Nucl Med. 2013;38(5):305-314. doi: 10.1097/RLU.0b013e3182867f3c [doi].
  • 25. Introduction and outline 23 15. Pokorny MR, de Rooij M, Duncan E, et al. Prospective study of diagnostic accuracy comparing prostate cancer detection by transrectal ultrasound-guided biopsy versus magnetic resonance (MR) imaging with subsequent MR-guided biopsy in men without previous prostate biopsies. Eur Urol. 2014;66(1):22-29. doi: 10.1016/j.eururo.2014.03.002 [doi]. 16. Siddiqui MM, Rais-Bahrami S, Turkbey B, et al. Comparison of MR/ultrasound fusion-guided biopsywithultrasound-guidedbiopsyforthediagnosisofprostatecancer.JAMA.2015;313(4):390- 397. doi: 10.1001/jama.2014.17942 [doi]. 17. Valerio M, Anele C, Charman SC, et al. Transperineal template prostate mapping biopsies: An evaluation of different protocols in detection of clinically significant prostate cancer. BJU Int. 2015. doi: 10.1111/bju.13306 [doi]. 18. Moore CM, Robertson NL, Arsanious N, et al. Image-guided prostate biopsy using magnetic resonance imaging-derived targets: A systematic review. Eur Urol. 2013;63(1):125-140. doi: 10.1016/j.eururo.2012.06.004 [doi]. 19. Wilt TJ, Brawer MK, Jones KM, et al. Radical prostatectomy versus observation for localised prostate cancer. N Engl J Med. 2012;367(3):203-213. doi: 10.1056/NEJMoa1113162 [doi]. 20. Bill-Axelson A, Holmberg L, Garmo H, et al. Radical prostatectomy or watchful waiting in early prostate cancer. N Engl J Med. 2014;370(10):932-942. doi: 10.1056/NEJMoa1311593 [doi]. 21. Schroder FH, Hugosson J, Roobol MJ, et al. Screening and prostate-cancer mortality in a randomizedeuropeanstudy.NEnglJMed.2009;360(13):1320-1328.doi:10.1056/NEJMoa0810084 [doi]. 22. Schroder FH, Hugosson J, Roobol MJ, et al. Prostate-cancer mortality at 11 years of follow-up. N Engl J Med. 2012;366(11):981-990. doi: 10.1056/NEJMoa1113135 [doi]. 23. Schroder FH, Hugosson J, Roobol MJ, et al. Screening and prostate cancer mortality: Results of the european randomised study of screening for prostate cancer (ERSPC) at 13 years of follow-up. Lancet. 2014;384(9959):2027-2035. doi: 10.1016/S0140-6736(14)60525-0 [doi]. 24. Andriole GL, Crawford ED, Grubb RL,3rd, et al. Prostate cancer screening in the randomized prostate, lung, colorectal, and ovarian cancer screening trial: Mortality results after 13 years of follow-up. J Natl Cancer Inst. 2012;104(2):125-132. doi: 10.1093/jnci/djr500 [doi]. 25. Dahm P, Neuberger M, Ilic D. Screening for prostate cancer: Shaping the debate on benefits and harms. Cochrane Database Syst Rev. 2013;9:ED000067. doi: org/10.1002/14651858.ED000067 [doi]. 26. Ilic D, Neuberger MM, Djulbegovic M, Dahm P. Screening for prostate cancer. Cochrane Database Syst Rev. 2013;1:CD004720. doi: 10.1002/14651858.CD004720.pub3 [doi]. 27. Resnick MJ, Koyama T, Fan KH, et al. Long-term functional outcomes after treatment for localised prostate cancer. N Engl J Med. 2013;368(5):436-445. doi: 10.1056/NEJMoa1209978 [doi]. 28. Klotz L, Zhang L, Lam A, Nam R, Mamedov A, Loblaw A. Clinical results of long-term follow-up of a large, active surveillance cohort with localised prostate cancer. J Clin Oncol. 2010;28(1):126- 131. doi: 10.1200/JCO.2009.24.2180 [doi].
  • 26. Introduction and outline 24 29. Grimm P, Billiet I, Bostwick D, et al. Comparative analysis of prostate-specific antigen free survival outcomes for patients with low, intermediate and high risk prostate cancer treatment by radical therapy. results from the prostate cancer results study group. BJU Int. 2012;109 Suppl 1:22-29. doi: 10.1111/j.1464-410X.2011.10827.x [doi]. 30. Cahlon O, Hunt M, Zelefsky MJ. Intensity-modulated radiation therapy: Supportive data for prostate cancer. Semin Radiat Oncol. 2008;18(1):48-57. doi: S1053-4296(07)00079-3 [pii]. 31. Guckenberger M, Flentje M. Intensity-modulated radiotherapy (IMRT) of localised prostate cancer: A review and future perspectives. Strahlenther Onkol. 2007;183(2):57-62. doi: 10.1007/ s00066-007-1631-3 [doi]. 32. Sheets NC, Goldin GH, Meyer AM, et al. Intensity-modulated radiation therapy, proton therapy, or conformal radiation therapy and morbidity and disease control in localised prostate cancer. JAMA. 2012;307(15):1611-1620. doi: 10.1001/jama.2012.460 [doi]. 33. Kim S, Shen S, Moore DF, et al. Late gastrointestinal toxicities following radiation therapy for prostate cancer. Eur Urol. 2011;60(5):908-916. doi: 10.1016/j.eururo.2011.05.052 [doi]. 34. Hoskin PJ, Rojas AM, Bownes PJ, Lowe GJ, Ostler PJ, Bryant L. Randomised trial of external beam radiotherapy alone or combined with high-dose-rate brachytherapy boost for localised prostate cancer. Radiother Oncol. 2012;103(2):217-222. doi: 10.1016/j.radonc.2012.01.007 [doi]. 35. Morton GC, Hoskin PJ. Brachytherapy: Current status and future strategies -- can high dose rate replace low dose rate and external beam radiotherapy? Clin Oncol (R Coll Radiol). 2013;25(8):474- 482. doi: 10.1016/j.clon.2013.04.009 [doi]. 36. DeSantis CE, Lin CC, Mariotto AB, et al. Cancer treatment and survivorship statistics, 2014. CA Cancer J Clin. 2014;64(4):252-271. doi: 10.3322/caac.21235 [doi]. 37. Morris WJ, Spadinger I, Keyes M, Hamm J, McKenzie M, Pickles T. Whole prostate D90 and V100: A dose-response analysis of 2000 consecutive (125)I monotherapy patients. Brachytherapy. 2014;13(1):32-41. doi: 10.1016/j.brachy.2013.08.006 [doi]. 38. Petereit DG, Frank SJ, Viswanathan AN, et al. Brachytherapy: Where has it gone? J Clin Oncol. 2015;33(9):980-982. doi: 10.1200/JCO.2014.59.8128 [doi]. 39. Hayes JH, Ollendorf DA, Pearson SD, et al. Observation versus initial treatment for men with localised, low-risk prostate cancer: A cost-effectiveness analysis. Ann Intern Med. 2013;158(12):853-860. doi: 10.7326/0003-4819-158-12-201306180-00002 [doi]. 40. Smith GD, Pickles T, Crook J, et al. Brachytherapy improves biochemical failure-free survival in low- and intermediate-risk prostate cancer compared with conventionally fractionated external beam radiation therapy: A propensity score matched analysis. Int J Radiat Oncol Biol Phys. 2015;91(3):505-516. doi: 10.1016/j.ijrobp.2014.11.018 [doi]. 41. Martin JM, Handorf EA, Kutikov A, et al. The rise and fall of prostate brachytherapy: Use of brachytherapy for the treatment of localised prostate cancer in the national cancer data base. Cancer. 2014;120(14):2114-2121. doi: 10.1002/cncr.28697 [doi]. 42. Nguyen PL, Chen MH, Zhang Y, et al. Updated results of magnetic resonance imaging guided partial prostate brachytherapy for favorable risk prostate cancer: Implications for focal therapy. J Urol. 2012;188(4):1151-1156. doi: 10.1016/j.juro.2012.06.010 [doi].
  • 27. Introduction and outline 25 43. Groenendaal G, Moman MR, Korporaal JG, et al. Validation of functional imaging with pathology for tumour delineation in the prostate. Radiother Oncol. 2010;94(2):145-150. doi: 10.1016/j. radonc.2009.12.034 [doi]. 44. Al-Qaisieh B, Mason J, Bownes P, et al. Dosimetry modeling for focal low-dose-rate prostate brachytherapy. Int J Radiat Oncol Biol Phys. 2015;92(4):787-793. doi: 10.1016/j.ijrobp.2015.02.043 [doi]. 45. Ahmed HU, Dickinson L, Charman S, et al. Focal ablation targeted to the index lesion in multifocal localised prostate cancer: A prospective development study. Eur Urol. 2015. doi: S0302- 2838(15)00073-1 [pii]. 46. Ahmed HU, Hindley RG, Dickinson L, et al. Focal therapy for localised unifocal and multifocal prostate cancer: A prospective development study. Lancet Oncol. 2012;13(6):622-632. doi: 10.1016/S1470-2045(12)70121-3 [doi]. 47. Bahn D, de Castro Abreu AL, Gill IS, et al. Focal cryotherapy for clinically unilateral, low- intermediate risk prostate cancer in 73 men with a median follow-up of 3.7 years. Eur Urol. 2012;62(1):55-63. doi: 10.1016/j.eururo.2012.03.006 [doi]. 48. Onik G, Vaughan D, Lotenfoe R, Dineen M, Brady J. "Male lumpectomy": Focal therapy for prostate cancer using cryoablation. Urology. 2007;70(6 Suppl):16-21. doi: 10.1016/j.urology.2007.06.001 [doi]. 49. Valerio M, Ahmed HU, Emberton M, et al. The role of focal therapy in the management of localised prostate cancer: A systematic review. Eur Urol. 2014;66(4):732-751. doi: 10.1016/j. eururo.2013.05.048 [doi]. 50. Giannarini G, Gandaglia G, Montorsi F, Briganti A. Will focal therapy remain only an attractive illusion for the primary treatment of prostate cancer? J Clin Oncol. 2014;32(13):1299-1301. doi: 10.1200/JCO.2013.54.8214 [doi]. 51. Valerio M, Emberton M, Ahmed HU. Focal therapy will become a standard option for selected men with localised prostate cancer. J Clin Oncol. 2014;32(32):3680-3681. doi: 10.1200/ JCO.2014.56.7792 [doi]. 52. Cahlon O, Zelefsky MJ, Shippy A, et al. Ultra-high dose (86.4 gy) IMRT for localised prostate cancer: Toxicity and biochemical outcomes. Int J Radiat Oncol Biol Phys. 2008;71(2):330-337. doi: 10.1016/j.ijrobp.2007.10.004 [doi]. 53. ZumstegZS,SprattDE,RomesserPB,etal.Thenaturalhistoryandpredictorsofoutcomefollowing biochemical relapse in the dose escalation era for prostate cancer patients undergoing definitive external beam radiotherapy. Eur Urol. 2015;67(6):1009-1016. doi: 10.1016/j.eururo.2014.09.028 [doi]. 54. Zelefsky MJ, Pei X, Chou JF, et al. Dose escalation for prostate cancer radiotherapy: Predictors of long-term biochemical tumour control and distant metastases-free survival outcomes. Eur Urol. 2011;60(6):1133-1139. doi: 10.1016/j.eururo.2011.08.029 [doi]. 55. Cellini N, Morganti AG, Mattiucci GC, et al. Analysis of intraprostatic failures in patients treated with hormonal therapy and radiotherapy: Implications for conformal therapy planning. Int J Radiat Oncol Biol Phys. 2002;53(3):595-599. doi: S0360301602027955 [pii].
  • 28. Introduction and outline 26 56. Pucar D, Hricak H, Shukla-Dave A, et al. Clinically significant prostate cancer local recurrence after radiation therapy occurs at the site of primary tumour: Magnetic resonance imaging and step-section pathology evidence. Int J Radiat Oncol Biol Phys. 2007;69(1):62-69. doi: S0360- 3016(07)00674-8 [pii]. 57. Arrayeh E, Westphalen AC, Kurhanewicz J, et al. Does local recurrence of prostate cancer after radiation therapy occur at the site of primary tumour? results of a longitudinal MRI and MRSI study. Int J Radiat Oncol Biol Phys. 2012;82(5):e787-93. doi: 10.1016/j.ijrobp.2011.11.030 [doi]. 58. Menard C, Iupati D, Publicover J, et al. MR-guided prostate biopsy for planning of focal salvage after radiation therapy. Radiology. 2015;274(1):181-191. doi: 10.1148/radiol.14122681 [doi]. 59. Cheng L, Cheville JC, Pisansky TM, et al. Prevalence and distribution of prostatic intraepithelial neoplasia in salvage radical prostatectomy specimens after radiation therapy. Am J Surg Pathol. 1999;23(7):803-808. 60. Huang WC, Kuroiwa K, Serio AM, et al. The anatomical and pathological characteristics of irradiated prostate cancers may influence the oncological efficacy of salvage ablative therapies. J Urol. 2007;177(4):1324-9; quiz 1591. doi: S0022-5347(06)03110-7 [pii]. 61. Leibovici D, Chiong E, Pisters LL, et al. Pathological characteristics of prostate cancer recurrence after radiation therapy: Implications for focal salvage therapy. J Urol. 2012;188(1):98-102. doi: 10.1016/j.juro.2012.02.2571 [doi]. 62. Andreoiu M, Cheng L. Multifocal prostate cancer: Biologic, prognostic, and therapeutic implications. Hum Pathol. 2010;41(6):781-793. doi: 10.1016/j.humpath.2010.02.011 [doi]. 63. Ahmed HU. The index lesion and the origin of prostate cancer. N Engl J Med. 2009;361(17):1704- 1706. doi: 10.1056/NEJMcibr0905562 [doi]. 64. Stamey TA, McNeal JM, Wise AM, Clayton JL. Secondary cancers in the prostate do not determine PSA biochemical failure in untreated men undergoing radical retropubic prostatectomy. Eur Urol. 2001;39 Suppl 4:22-23. doi: 52577 [pii]. 65. Wise AM, Stamey TA, McNeal JE, Clayton JL. Morphologic and clinical significance of multifocal prostate cancers in radical prostatectomy specimens. Urology. 2002;60(2):264-269. doi: S0090429502017284 [pii]. 66. Liu W, Laitinen S, Khan S, et al. Copy number analysis indicates monoclonal origin of lethal metastatic prostate cancer. Nat Med. 2009;15(5):559-565. doi: 10.1038/nm.1944 [doi]. 67. Haffner MC, Mosbruger T, Esopi DM, et al. Tracking the clonal origin of lethal prostate cancer. J Clin Invest. 2013;123(11):4918-4922. doi: 10.1172/JCI70354 [doi]. 68. Ahmed HU, Pendse D, Illing R, Allen C, van der Meulen JH, Emberton M. Will focal therapy become a standard of care for men with localised prostate cancer? Nat Clin Pract Oncol. 2007;4(11):632-642. doi: ncponc0959 [pii]. 69. Eggener SE, Scardino PT, Carroll PR, et al. Focal therapy for localised prostate cancer: A critical appraisal of rationale and modalities. J Urol. 2007;178(6):2260-2267. doi: S0022-5347(07)02162-3 [pii].
  • 29. Introduction and outline 27 70. Kasivisvanathan V, Emberton M, Ahmed HU. Focal therapy for prostate cancer: Rationale and treatment opportunities. Clin Oncol (R Coll Radiol). 2013;25(8):461-473. doi: 10.1016/j. clon.2013.05.002 [doi]. 71. Klotz L, Emberton M. Management of low risk prostate cancer-active surveillance and focal therapy. Nat Rev Clin Oncol. 2014;11(6):324-334. doi: 10.1038/nrclinonc.2014.73 [doi]. 72. Donaldson IA, Alonzi R, Barratt D, et al. Focal therapy: Patients, interventions, and outcomes--a report from a consensus meeting. Eur Urol. 2015;67(4):771-777. doi: 10.1016/j.eururo.2014.09.018 [doi]. 73. van den Bos W, Muller BG, Ahmed H, et al. Focal therapy in prostate cancer: International multidisciplinary consensus on trial design. Eur Urol. 2014;65(6):1078-1083. doi: 10.1016/j. eururo.2014.01.001 [doi]. 74. Lips IM, van der Heide UA, Haustermans K, et al. Single blind randomized phase III trial to investigate the benefit of a focal lesion ablative microboost in prostate cancer (FLAME-trial): Study protocol for a randomized controlled trial. Trials. 2011;12:255-6215-12-255. doi: 10.1186/1745- 6215-12-255 [doi]. 75. Ahmed HU. Introduction--targeting the lesion, not the organ. Urol Oncol. 2014;32(6):901-902. doi: 10.1016/j.urolonc.2014.06.011 [doi]. 76. Chade DC, Shariat SF, Cronin AM, et al. Salvage radical prostatectomy for radiation-recurrent prostate cancer: A multi-institutional collaboration. Eur Urol. 2011;60(2):205-210. doi: 10.1016/j. eururo.2011.03.011 [doi]. 77. Chade DC, Eastham J, Graefen M, et al. Cancer control and functional outcomes of salvage radical prostatectomy for radiation-recurrent prostate cancer: A systematic review of the literature. Eur Urol. 2012;61(5):961-971. doi: 10.1016/j.eururo.2012.01.022 [doi]. 78. Paparel P, Cronin AM, Savage C, Scardino PT, Eastham JA. Oncologic outcome and patterns of recurrence after salvage radical prostatectomy. Eur Urol. 2009;55(2):404-410. doi: 10.1016/j. eururo.2008.07.007 [doi]. 79. Wenske S, Quarrier S, Katz AE. Salvage cryosurgery of the prostate for failure after primary radiotherapy or cryosurgery: Long-term clinical, functional, and oncologic outcomes in a large cohort at a tertiary referral centre. Eur Urol. 2013;64(1):1-7. doi: 10.1016/j.eururo.2012.07.008 [doi]. 80. Williams AK, Martinez CH, Lu C, Ng CK, Pautler SE, Chin JL. Disease-free survival following salvage cryotherapy for biopsy-proven radio-recurrent prostate cancer. Eur Urol. 2011;60(3):405- 410. doi: 10.1016/j.eururo.2010.12.012 [doi]. 81. Spiess PE, Katz AE, Chin JL, et al. A pretreatment nomogram predicting biochemical failure after salvage cryotherapy for locally recurrent prostate cancer. BJU Int. 2010;106(2):194-198. doi: 10.1111/j.1464-410X.2009.09078.x [doi]. 82. Crouzet S, Murat FJ, Pommier P, et al. Locally recurrent prostate cancer after initial radiation therapy: Early salvage high-intensity focused ultrasound improves oncologic outcomes. Radiother Oncol. 2012;105(2):198-202. doi: 10.1016/j.radonc.2012.09.014 [doi].
  • 30. Introduction and outline 28 83. Murat FJ, Poissonnier L, Rabilloud M, et al. Mid-term results demonstrate salvage high- intensity focused ultrasound (HIFU) as an effective and acceptably morbid salvage treatment option for locally radiorecurrent prostate cancer. Eur Urol. 2009;55(3):640-647. doi: 10.1016/j. eururo.2008.04.091 [doi]. 84. Chen CP, Weinberg V, Shinohara K, et al. Salvage HDR brachytherapy for recurrent prostate cancer after previous definitive radiation therapy: 5-year outcomes. Int J Radiat Oncol Biol Phys. 2013;86(2):324-329. doi: 10.1016/j.ijrobp.2013.01.027 [doi]. 85. Grado GL, Collins JM, Kriegshauser JS, et al. Salvage brachytherapy for localised prostate cancer after radiotherapy failure. Urology. 1999;53(1):2-10. doi: S0090-4295(98)00492-0 [pii]. 86. Moman MR, van der Poel HG, Battermann JJ, Moerland MA, van Vulpen M. Treatment outcome and toxicity after salvage 125-I implantation for prostate cancer recurrences after primary 125-I implantation and external beam radiotherapy. Brachytherapy. 2010;9(2):119-125. doi: 10.1016/j. brachy.2009.06.007 [doi]. 87. Burri RJ, Stone NN, Unger P, Stock RG. Long-term outcome and toxicity of salvage brachytherapy for local failure after initial radiotherapy for prostate cancer. Int J Radiat Oncol Biol Phys. 2010;77(5):1338-1344. doi: 10.1016/j.ijrobp.2009.06.061 [doi]. 88. Punnen S, Cooperberg MR, D'Amico AV, et al. Management of biochemical recurrence after primary treatment of prostate cancer: A systematic review of the literature. Eur Urol. 2013;64(6):905-915. doi: 10.1016/j.eururo.2013.05.025 [doi]. 89. Parekh A, Graham PL, Nguyen PL. Cancer control and complications of salvage local therapy after failure of radiotherapy for prostate cancer: A systematic review. Semin Radiat Oncol. 2013;23(3):222-234. doi: 10.1016/j.semradonc.2013.01.006 [doi]. 90. Kimura M, Mouraviev V, Tsivian M, Mayes JM, Satoh T, Polascik TJ. Current salvage methods for recurrent prostate cancer after failure of primary radiotherapy. BJU Int. 2010;105(2):191-201. doi: 10.1111/j.1464-410X.2009.08715.x [doi]. 91. Nguyen PL, D'Amico AV, Lee AK, Suh WW. Patient selection, cancer control, and complications after salvage local therapy for postradiation prostate-specific antigen failure: A systematic review of the literature. Cancer. 2007;110(7):1417-1428. doi: 10.1002/cncr.22941 [doi]. 92. Heidenreich A, Bastian PJ, Bellmunt J, et al. EAU guidelines on prostate cancer. part II: Treatment of advanced, relapsing, and castration-resistant prostate cancer. Eur Urol. 2014;65(2):467-479. doi: 10.1016/j.eururo.2013.11.002 [doi]. 93. Nguyen PL, Alibhai SM, Basaria S, et al. Adverse effects of androgen deprivation therapy and strategies to mitigate them. Eur Urol. 2015;67(5):825-836. doi: 10.1016/j.eururo.2014.07.010 [doi]. 94. Tran H, Kwok J, Pickles T, Tyldesley S, Black PC. Underutilization of local salvage therapy after radiation therapy for prostate cancer. Urol Oncol. 2014;32(5):701-706. doi: 10.1016/j. urolonc.2013.12.014 [doi]. 95. Agarwal PK, Sadetsky N, Konety BR, Resnick MI, Carroll PR, Cancer of the Prostate Strategic Urological Research Endeavor (CaPSURE). Treatment failure after primary and salvage therapy for prostate cancer: Likelihood, patterns of care, and outcomes. Cancer. 2008;112(2):307-314. doi: 10.1002/cncr.23161 [doi].
  • 31. Introduction and outline 29 96. Alongi F, De Bari B, Campostrini F, et al. Salvage therapy of intraprostatic failure after radical external-beamradiotherapyforprostatecancer:Areview.CritRevOncolHematol.2013;88(3):550- 563. doi: 10.1016/j.critrevonc.2013.07.009 [doi]. 97. Marks LB, Carroll PR, Dugan TC, Anscher MS. The response of the urinary bladder, urethra, and ureter to radiation and chemotherapy. Int J Radiat Oncol Biol Phys. 1995;31(5):1257-1280. doi: 0360-3016(94)00431-J [pii]. 98. Peters M, Moman MR, van der Poel HG, et al. Patterns of outcome and toxicity after salvage prostatectomy, salvage cryosurgery and salvage brachytherapy for prostate cancer recurrences after radiation therapy: A multi-center experience and literature review. World J Urol. 2013;31(2):403- 409. doi: 10.1007/s00345-012-0928-8 [doi]. 99. Crehange G, Roach M,3rd, Martin E, et al. Salvage reirradiation for locoregional failure after radiation therapy for prostate cancer: Who, when, where and how? Cancer Radiother. 2014;18(5- 6):524-534. doi: 10.1016/j.canrad.2014.07.153 [doi]. 100. Bomers JG, Yakar D, Overduin CG, et al. MR imaging-guided focal cryoablation in patients with recurrent prostate cancer. Radiology. 2013;268(2):451-460. doi: 10.1148/radiol.13121291 [doi]. 101. de Castro Abreu AL, Bahn D, Leslie S, et al. Salvage focal and salvage total cryoablation for locally recurrent prostate cancer after primary radiation therapy. BJU Int. 2013;112(3):298-307. doi: 10.1111/bju.12151 [doi]. 102. Eisenberg ML, Shinohara K. Partial salvage cryoablation of the prostate for recurrent prostate cancer after radiotherapy failure. Urology. 2008;72(6):1315-1318. doi: 10.1016/j. urology.2008.03.040 [doi]. 103. Li YH, Elshafei A, Agarwal G, Ruckle H, Powsang J, Jones JS. Salvage focal prostate cryoablation for locally recurrent prostate cancer after radiotherapy: Initial results from the cryo on-line data registry. Prostate. 2015;75(1):1-7. doi: 10.1002/pros.22881 [doi]. 104. Ahmed HU, Cathcart P, McCartan N, et al. Focal salvage therapy for localised prostate cancer recurrence after external beam radiotherapy: A pilot study. Cancer. 2012;118(17):4148-4155. doi: 10.1002/cncr.27394 [doi]. 105. Baco E, Gelet A, Crouzet S, et al. Hemi salvage high-intensity focused ultrasound (HIFU) in unilateral radiorecurrent prostate cancer: A prospective two-centre study. BJU Int. 2014;114(4):532-540. doi: 10.1111/bju.12545 [doi]. 106. Hsu CC, Hsu H, Pickett B, et al. Feasibility of MR imaging/MR spectroscopy-planned focal partial salvage permanent prostate implant (PPI) for localised recurrence after initial PPI for prostate cancer. Int J Radiat Oncol Biol Phys. 2013;85(2):370-377. doi: 10.1016/j.ijrobp.2012.04.028 [doi]. 107. Peters M, Maenhout M, van der Voort van Zyp JRN, et al. Focal salvage iodine-125 brachytherapy for prostate cancer recurrences after primary radiotherapy: A retrospective study regarding toxicity, biochemical outcome and quality of life. Radiother Oncol. 2014;112(1):77-82. doi: 10.1016/j.radonc.2014.06.013 [doi]. 108. Nguyen PL, Chen MH, D'Amico AV, et al. Magnetic resonance image-guided salvage brachytherapy after radiation in select men who initially presented with favorable-risk prostate cancer: A prospective phase 2 study. Cancer. 2007;110(7):1485-1492. doi: 10.1002/cncr.22934 [doi].
  • 32. Introduction and outline 30 109. Crook JM, Potters L, Stock RG, Zelefsky MJ. Critical organ dosimetry in permanent seed prostate brachytherapy: Defining the organs at risk. Brachytherapy. 2005;4(3):186-194. doi: S1538- 4721(05)00108-X [pii]. 110. Davis BJ, Horwitz EM, Lee WR, et al. American brachytherapy society consensus guidelines for transrectal ultrasound-guided permanent prostate brachytherapy. Brachytherapy. 2012;11(1):6- 19. doi: 10.1016/j.brachy.2011.07.005 [doi]. 111. Price JG, Stone NN, Stock RG. Predictive factors and management of rectal bleeding side effects following prostate cancer brachytherapy. Int J Radiat Oncol Biol Phys. 2013;86(5):842-847. doi: 10.1016/j.ijrobp.2013.04.033 [doi]. 112. Hathout L, Folkert MR, Kollmeier MA, Yamada Y, Cohen GN, Zelefsky MJ. Dose to the bladder neck is the most important predictor for acute and late toxicity after low-dose-rate prostate brachytherapy: Implications for establishing new dose constraints for treatment planning. Int J Radiat Oncol Biol Phys. 2014;90(2):312-319. doi: 10.1016/j.ijrobp.2014.06.031 [doi]. 113. Salembier C, Lavagnini P, Nickers P, et al. Tumour and target volumes in permanent prostate brachytherapy: A supplement to the ESTRO/EAU/EORTC recommendations on prostate brachytherapy. Radiother Oncol. 2007;83(1):3-10. doi: S0167-8140(07)00028-X [pii]. 114. Snyder KM, Stock RG, Hong SM, Lo YC, Stone NN. Defining the risk of developing grade 2 proctitis following 125I prostate brachytherapy using a rectal dose-volume histogram analysis. Int J Radiat Oncol Biol Phys. 2001;50(2):335-341. doi: S0360-3016(01)01442-0 [pii]. 115. Henriquez I, Sancho G, Hervas A, et al. Salvage brachytherapy in prostate local recurrence after radiation therapy: Predicting factors for control and toxicity. Radiat Oncol. 2014;9:102-717X-9- 102. doi: 10.1186/1748-717X-9-102 [doi].
  • 33.
  • 34. Max Peters1 Maaike R. Moman1 Henk G. van der Poel2 Henk Vergunst3 Igle Jan de Jong4 Peter L.M. Vijverberg5 Jan J. Battermann1 Simon Horenblas2 Marco van Vulpen1 1 Department of Radiation Oncology, University Medical Centre Utrecht, Utrecht, The Netherlands 2 Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands 3 Department of Urology, Canisius-Wilhelmina Hospital, Nijmegen, The Netherlands 4 Department of Urology, University Medical Centre Groningen, Groningen, The Netherlands 5 Department of Urology, St. Antonius Hospital, Nieuwegein, The Netherlands This work was published previously in: World Journal of Urology (2013) 31:403–409. DOI 10.1007/s00345-012-0928-8
  • 35. Patterns of outcome and toxicity after salvage prostatectomy, salvage cryosurgery and salvage Iodine-125 brachytherapy for prostate cancer recurrences after radiation therapy: a multi-centre experience and literature comparison Chapter 1
  • 36. Chapter1 Chapter 1 34 Abstract Purpose Current salvage treatments for recurrent prostate cancer after primary radiation therapy include radical prostatectomy, cryosurgery and brachytherapy. Because toxicity and failure rates are considerable, salvage treatments are not commonly performed. As most centres perform only one preferred salvage technique, the literature only describes single-centre outcomes from a particular salvage technique with a limited number of patients. In this overview, five high-volume Dutch centres describe their toxicity and outcome data using different salvage techniques: salvage radical prostatectomy, salvage Iodine-125 (I-125) brachytherapy and salvage cryosurgery. This provides insight on how salvage is performed in clinical practice in the Netherlands. Methods A total of 129 patients from five different centres in the Netherlands were retrospectively analysed. Biochemical failure (BF) was defined as PSA >0.1 ng/ml for the salvage prostatectomy group (n=44) and PSA nadir + 2.0 ng/ml (Phoenix definition) for the salvage cryosurgery (n=54) and salvage I-125 brachytherapy group (n=31). Toxicity was scored according to the Common Terminology Criteria for Adverse events (CTCAE v3.0). Results BF occurred in 25 (81%) patients in the brachytherapy group after mean follow-up of 29 (SD 24) months, in 29 patients (66%) patients in the prostatectomy group after mean follow-up of 22 (SD 25) months and in 33 patients (61%) patients in the cryosurgery group after mean follow-up of 14 (SD 11) months. Severe (≥grade 3) genitourinary and gastrointestinal toxicity was observed in up to 30% of patients in all three cohorts. Conclusion This overview shows clinical practice of prostate cancer salvage in the Netherlands. Significant failure and toxicity rates are observed, regardless of salvage technique. Patients should be selected with great care before offering these salvage treatment strategies.
  • 37. Chapter1 Outcomes of whole-gland salvage in the Netherlands 35 Introduction Asignificantproportionofprostatecancerpatientstreatedwithprimaryradiotherapy develops a recurrence. Estimations of risks exceeding 50% have been presented for the highest risk groups1-4 . The recurrence risk is dependent on various risk factors such as initial PSA level (iPSA), PSA doubling time (PSADT), Gleason score and PSA nadir after initial treatment5 . A significant number of patients are thought to harbour organ-confined disease6,7 . It seems local recurrences are particularly situated at the position of the originally largest tumour, or index lesion8 . The currently dominantly applied curative salvage treatments for these recurrences are salvage brachytherapy, prostatectomyandcryosurgery.Unfortunately,thesesalvagetreatmentsareassociated with high failure and severe toxicity rates9,10 . As most centres perform one preferred salvage technique, the literature only describes single-centre outcomes with a limited number of patients. In this overview, five high-volume Dutch centres describe their outcomes using different salvage techniques. This provides a view on how salvage is performed in clinical practice in the Netherlands. Methods and materials Treatment outcome and toxicity were recorded retrospectively for the salvage treatments given: salvage radical prostatectomy (SRP), salvage cryosurgery (SCS) and salvage Iodine-125 (I-125) implantation/brachytherapy (SBT). Primary treatments were external beam radiation therapy or I-125 implantation. All men showed PSA failure afterward, and recurrences were confirmed by transrectal ultrasound (TRUS) guided biopsies. Furthermore, a bone scan and CT-scan were made to exclude any possible distant or lymph node metastases. A PET-CT scan was done in the more recent patients, but this imaging modality is not general practice in the Netherlands. Patients were selected for salvage treatment based on a life expectancy of at least 10 years, and all were counselled with respect to expected toxicity and cancer control based on the experience of the institute. Salvage procedures All SRP procedures were performed at the Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AVL), Amsterdam, via a perineal approach. Since 2003, 44 patients underwent SRP. A pelvic lymph node dissection was performed before SRP in all patients, to exclude metastases. A median of 13 lymph nodes were analysed.Theprocedureisdescribedelsewhereinmoredetail11 .Nonervesparingwas attempted. Five patients received hormonal treatment ≤3 months prior to surgery. SBT was performed by I-125 seed implantation at the University Medical Centre
  • 38. Chapter1 Chapter 1 36 Utrecht (UMCU). Since 1993, 31 patients have been treated with permanent I-125 seed implants, via a transperineal route under ultrasound guidance. The standard target dose was ≥145 Gy12 . The technique has been described previously13 . Only patients with a prostate volume ≤50 cc were eligible for implantation. A total of five patients received hormonal treatment three months prior to salvage in order to decrease the prostatic volume. For the implants and the doses received by the surrounding structures (rectum, bladder, urethra), the following criteria were used: the prostatic V100 (the volume of the prostate that receives 100% of the prescribed dose) ≥95%, the prostatic D90 (minimal dose to 90% of the prostatic volume) ≥145 Gy, the rectal D2cc (the minimal dose received by the maximally irradiated 2cc of the structure) ≤145 Gy and the D10 (the minimal dose to 10% of the structure) of the prostatic urethra ≤150% of 145 Gy12,14 . Cryosurgery was performed at the Canisius-Wilhelmina Hospital (CWH), Nijmegen, the University Medical Centre Groningen (UMCG) and the Sint Antonius Hospital (St.AH), Nieuwegein. Since 2007, 54 patients underwent salvage cryosurgery. Cryosurgery was performed with Seednet (prostate <35 cc) and Icerod needles (prostate >35 cc) inserted transperineally through a brachytherapy template. The 17 Gauge needles were placed 1 cm apart with TRUS visualisation. A transurethral warming catheter was inserted to protect the urethra. A suprapubic catheter was left in situ for two weeks, in combination with antibiotic prophylaxis. Patients were treated with two cycles of freezing and thawing. Thermosensors in the midportion of the prostate gland ensured that the required temperature of -40 0C was reached for effective cell killing. Biochemical failure and toxicity BF for patients after SRP was defined as a serum PSA level >0.1 ng/ml. For SBT and SCS, failure according to the Phoenix and ASTRO definition was assessed: PSA nadir + 2 ng/ml (Phoenix) and three consecutive rises in PSA with backdating of the moment of failure to midway between the nadir after treatment and the first rise (ASTRO)15,16 . Toxicity was scored from file records based on structured interviewsatdifferenttimeintervalsaftersalvagetreatment.Toimproveuniformity, the toxicity from each clinic was retrospectively converted to a grade using the Common Terminology Criteria for Adverse Events, version 3.0 (CTCAE v3.0)17 . No subdivision into acute and late toxicity was made for this analysis. Statistical analysis Descriptive statistics and Kaplan–Meier survival analyses for freedom from biochemical failure (FFF) were performed with the Statistical Package for the
  • 39. Chapter1 Outcomes of whole-gland salvage in the Netherlands 37 Social Sciences, version 17.0 (SPSS, Chicago, IL, USA). Differences in Kaplan- Meier FFF estimates were tested with the log-rank test, with statistical significance set at p ≤0.05. Toxicity is presented as the proportion of patients suffering from genitourinary (GU) or gastrointestinal (GI) morbidity and erectile dysfunction (ED). Results A total of 129 patients were included. Various baseline characteristics differed betweenthegroups,inparticularage,iPSA,clinicaltumourstageandfollow-uptime (Table 1). SBT and SCS patients were older at the time of salvage than SRP patients (69 and 70 years vs. 65 years). The iPSA was higher than 20 ng/ml in 35% of the SBT group and 30% of the SCS group, versus 5% in the SRP group. Primary cT3 tumours were19%inSBTpatients,32%inSRPpatientsand39%inSCSpatients.Meanfollow- up in the SCS group was 3 years, in comparison with 5 and 9 years for SRP and SBT, respectively.Resultsregardingbiochemicalresponse,mortalityandsevere(grade3) toxicity are presented in table 2. Figure 1 shows the Kaplan–Meier curve for the FFF. In the SRP group, 29 (66%) patients suffered from BF after mean 22 months (SD 25 months). Grade 1 or 2 GU toxicity occurred in 24 (55%) patients, and grade 3 toxicity occurred in 10 (23%) patients. Grade 1 or 2 GI toxicity occurred in three (7%) patients, and grade 3 (fistulas) occurred in four (9%) patients. Until now, five (11%)patientshavediedduetoprostatecancer.EDwasobservedin86%ofpatients. After I-125 implantation, 25 (81%) patients suffered from BF after mean 29 months (SD 24 months). Grade 1 or 2 GU toxicity occurred in 17 (55%) patients, and grade 3 occurred in seven (23%) patients, predominantly urinary incontinence and obstruction due to swelling of the prostate. Grade 1 and 2 GI toxicity occurred in 16 (52%) patients, and grade 3 occurred in two (6%) patients. Twelve (39%) patients have died due to prostate cancer. ED was present in 45% of patients. After a mean 14 months after SCS, 33 (61%) patients suffered from BF after mean 14 months (SD 11 months). Grade 1 and 2 GU toxicity occurred in 30 (56%) patients, and grade 3 occurred in 12 (22%) patients. Urinary incontinence, obstruction and urinary tract infection were frequent complications. Perineal pain was also a common complaint. Grade 1 and 2 GI toxicity occurred in seven (13%) patients, and grade 3 occurred in four (7%) patients (predominantly fistula formation between the rectum and urethra). ED was observed in 50 (93%) patients. Twenty- four (44%) patients had pre-existent ED. No patients have died due to prostate cancer–related events.
  • 40. Chapter1 Chapter 1 38 Table 1: Baseline characteristics of the study population, per salvage treatment group Prostatectomy I-125 Cryosurgery N 44 31 54 Mean (SD) age at primary diagnosis, years 60 (6) 64 (5) 68 (6) Mean (SD) age at salvage treatment, years 65 (6) 69 (5) 70 (6) Primary tumour stage cT1 cT2 cT3 10 (23%) 20 (46%) 14 (32%) 5 (16%) 20 (65%) 6 (19%) 16 (30%) 17 (31%) 21 (39%) Gleason score Gleason 2 - 6 Gleason 7 Gleason 8 – 10 Unknown 18 (41%) 19 (43%) 7 (16%) 13 (42%) 13 (42%) 2 (6%) 3 (10%) 14 (26%) 16 (30%) 21 (39%) 3 (5%) Primary treatment EBRT HDR I-125 IMRT 31 (70%) 2 (5%) 11 (25%) 0 20 (64%) 0 11 (36%) 0 43 (80%) 0 4 (7%) 7 (13) Initial PSA 0 – 10 ng/ml > 10 – 20 ng/ml > 20 ng/ml Unknown 12 (27%) 29 (66%) 2 (5%) 1 (2%) 5 (16%) 7 (23%) 11 (35%) 8 (26%) 16 (30%) 15 (27%) 16 (30%) 7 (13%) PSA before salvage, ng/ml 0 – 10 ng/ml > 10 – 20 ng/ml > 20 ng/ml Unknown 24 (55%) 18 (41%) 2 (5%) 17 (55%) 8 (26%) 5 (16%) 1 (3%) 42 (78%) 9 (17%) 3 (5%) 0 Mean (SD) PSADT before salvage, months 13 (8) 13 (9) 15 (5) Mean (SD) prostate volume before salvage, cm3 28 (2) 24 (10) 21 (8) Number of patients treated with hormonal therapy <3 months before salvage 5 (11%) 5 (16%) 8 (15%) Abbreviations: I-125=Iodine-125; SD=standard deviation; EBRT=external beam radiotherapy; HDR=high dose rate radiotherapy; IMRT=Intensity Modulated Radiotherapy; PSADT=PSA doubling time.
  • 41. Chapter1 Outcomes of whole-gland salvage in the Netherlands 39 Table 2: Biochemical failure, mortality and toxicity rates after salvage, per salvage treatment group Prostatectomy I-125 Cryosurgery N 44 31 54 Mean (SD) FU after salvage, years 5 (4) 9 (3) 3 (0,3) Mean (SD) PSA nadir, ng/ml 0.2 (0.8) 4.7 (6.6) 2 (3) Mean (SD) time to nadir, months 1.5 10 (9) 2 (2) Patients with biochemical failure† Phoenix ASTRO PSA >0.1 ng/ml NA NA 29 (66%) 25 (81%) 17 (55%) NA 33 (61%) 32 (59%) NA Mean (SD) time to failure, months Phoenix ASTRO PSA >0.1 ng/ml NA NA 22 (25) 29 (24) 41 (25) NA 14 (11) 16 (9) NA Patients (%) with severe GU toxicity, CTCAE Grade 3 10 (23%) 7 (23%) 12 (22%) Patients (%) with severe GI toxicity, CTCAE Grade 3 4 (9%) 2 (6%) 4 (7%) Death Yes, PCa-specific Yes, other cause 5 (11%) 7 (16%) 12 (39%) 2 (6%) 0 (0%) 5 (9%) Abbreviations: I-125=Iodine-125; FU=follow up; SD=standard deviation; ASTRO=American Society for Therapeutic Radiology and Oncology; GU=genitourinary; GI=gastrointestinal; PCa=prostate cancer; CTCAE=Common Terminology Criteria for Adverse Events v3.0; NA=Not applicable. † Proportion of patients with biochemical failure, as defined by 2 different criteria. Definitions of biochemical failure: Phoenix=PSA nadir plus 2 ng/ml, ASTRO=3 consecutive rises in PSA backdating the moment of failure to midway between the nadir after treatment and the first rise.
  • 42. Chapter1 Chapter 1 40 Discussion This paper shows how salvage for radiorecurrent prostate cancer is performed in the Netherland and reports on the current cancer control outcomes and toxicity. Biochemical failure occurs often, with an estimated FFF of maximally 27% after 5years.Toxicitywasalsofrequent,withapproximately30%ofpatientsexperiencing a grade 3 or higher complication of the genitourinary and gastrointestinal tract combined.Intheliterature,awidevariationofpapersdescribestreatmentoutcomes and toxicity after different salvage modalities, preceded by different primary radiation therapy modalities and schedules. To our knowledge, no previous article has presented outcomes of three different salvage treatment modalities from one population. A comparison with the literature is summarised in table 3 (FFF) and table 4 (toxicity). Figure 1: Kaplan-Meier curve showing the freedom from biochemical failure following salvage radical prostatectomy, salvage Iodine-125 implantation and salvage cryotherapy (Log rank: p=0.42). The numbers at risk at 5 years were 7 for salvage radical prostatectomy, 5 for Iodine-125 brachytherapy and 6 (at three years) for salvage cryosurgery.
  • 43. Chapter1 Outcomes of whole-gland salvage in the Netherlands 41 Table 3: Comparison with results from the literature – freedom from biochemical failure. Salvage procedure No. studies [References] N Follow up FFF (%) Follow up present study FFF present study (%) Radical prostatectomy 4[10,11,19,27,28] 337 5 years 31 – 71 5 years 27 I-125 implantation 2[10,21,29] 66 5 years 34 – 53 5 years 19 Cryosurgery 3[10,22,24,30] 597 5 years 40 – 55 3 years 19 Abbreviations: FFF=freedom from biochemical failure; I-125=Iodine-125. Table 4: Comparison with results from the literature – toxicity Salvage procedure Radical prostatectomy I-125 implantation Cryosurgery Literature No. studies [References] 4[10,11,19,27,31] 2[10,21,29] 2[10,23,30] N 335 66 397 % GU toxicity 20-68% incontinence 22-41% bladder neck stricture 0-47% grade 3-4 4-83% incontinence 7-55% bladder neck stricture / retention % GI toxicity 2-7% rectal injury 0-24% grade 3-4 6-37% perineal pain 1-11% fistula Erectile dysfunction 72 %—Nearly uniform NA 72–86 % Present study % GU toxicity 23% grade 3 23% grade 3 22% grade 3 % GI toxicity 9% grade 3 6% grade 3 7% grade 3 Erectile dysfunction 86% 45% 93%* Abbreviations: I-125=Iodine-125; GU=genitourinary; GI=gastrointestinal. NA=Not available * 44% of patients had pre-existent erectile dysfunction. However,thiscomparisonishamperedbythesmallanddivergentstudypopulations and the different definitions for BF. Although the current study population is not equivalent regarding patient and tumour characteristics and follow-up, it might be possible to make a crude comparison regarding toxicity. Grade 3 GU and GI toxicity occurredin32%ofSRPpatients,29%ofSBTpatientsand30%ofSCSpatients.These toxicity rates are approximately equivalent to results as found in the literature9,10 . The5-yearFFFwas27%followingSRPand19%followingSBT(Phoenixdefinition). After 36 months, the FFF following cryosurgery was 19%. These figures are lower
  • 44. Chapter1 Chapter 1 42 compared to those found in the literature (Table 3 & 4 and figure 1). This could be due to incomplete salvage procedures, but dissemination before salvage is more likely. Especially the cryosurgery group contained high risk cases with cT3 and Gleason8–10tumours.Theliteratureshowsthatthepre-salvagePSAconcentration, PSADT after primary radiation treatment and the tumour stage and grade are important in predicting biochemical recurrences after salvage procedures9-11,18 . In one SRP study, patients with a pre-salvage PSA <2 ng/ml and PSADT >12 months showed better biochemical disease free survival (bDFS) rates than patient with PSA >2 ng/ml and PSADT <12 months: >80% bDFS after 80 months in the most favourable risk group compared with 0% bDFS after 20 months in the group with the highest risk factors11 . Other SRP data indicate more favourable results for primary T2 tumours in comparison with T3 tumours (100% bDFS after 5 years in T2 tumours compared to 35% in T3 and 0% in N+ tumours)19 . In addition, the tumour stage (T3/T4) was found to be an independent predictor of cancer specific survival in a large series of SRP patients after multivariable analysis (n=199)18 . In SBT, patients with pre-salvage Gleason scores ≥7 and PSADT <10 months show poorer biochemical control rates in multivariable analysis10,20 . Possibly, a pre- salvage PSA >10 ng/ml also leads to poorer cancer control outcomes21 . For the SCS group, these risk factors for salvage treatment failure have also been recognised. Patients with a pre-salvage PSA >10 ng/ml, Gleason scores ≥8 before primary treatment and clinical T3/T4 tumours show decreased biochemical control rates in comparison with patients without these risk factors9,10,22-24 . For an accurate selection of patients likely to benefit from salvage therapy, these risk factors have to be incorporated in salvage protocols. The general absence of these selection criteria in these cohorts has probably contributed to the fairly low cancer control rates observed. These pre-salvage characteristics would need to be researched with the use of adequate prediction modelling to aid patient selection in the future. The retrospective nature of this analysis yields several additional limitations. First, the study populations are different at baseline, and therefore especially the results regarding cancer control are difficult to compare between the three salvage modalities. Also, the FFF rates depend on the criteria used to define BF. For SBT and SCS, the Phoenix definition was used, because the ASTRO definition is more prone to bias because of variable follow-up intervals. The ASTRO definition furthermore has a false-positive rate of approximately 20% after the use of hormonal therapy, versus5%associatedwiththePhoenixdefinition15,16 .Further,becausethegroupsare limited, analysis of risk factors in a multivariable model was not perfomed. Finally, the use of hormonal treatment was not standardised as well, further hampering
  • 45. Chapter1 Outcomes of whole-gland salvage in the Netherlands 43 comparisons between groups. The primary objective of this study, however, was not to provide an exhaustive comparison regarding different salvage modalities, but to show the current clinical practice in the Netherlands. This can provide an indication of what aspects need to be considered when offering salvage. Patients should be intensely counselled regarding the high toxicity rates. These numbers might even be underestimated due to the retrospective nature of the analysis, creating the possibilitythatsalvagecanbeevenmoredetrimentalthanobservedinthesecohorts. In addition, cancer control outcomes are suboptimal, which is possibly related to inaccurate patient selection. In the SBT group, treatment goes back as far as 1993. The patients selected in this period often had high-risk characteristics that would exclude them from salvage treatment today. It is possible that especially this lack of proper patient selection has negatively influenced the FFF. For future research, it is important to pay further attention to patient selection. The most important aspect is clearly defining local or distant disease at the time of BF after primary radiotherapy. The PSA value is one of the most important diagnostic and follow-up measurement available today, but is insufficient to assess disease progression. A rising PSA level after primary radiotherapy can have several possible origins, including lymph-node or distant metastases. Furthermore, the currently applied Phoenix definition has a false positive rate of 5%16 . Better imaging techniques could contribute to a better selection of patients with a local (organ-confined) recurrence and exclude patients with systemic disease25,26 . Prediction models taking into account several pre-salvage characteristicscouldfurtheraidpatientselection.Thesemodelsincombinationwith accurate diagnostic modalities should be able to lead to a better patient selection for salvage treatment in the future, thereby improving cancer control outcomes. Lastly, with better assessment of localised disease, focal salvage directed only at the recurrent tumour area might be able to decrease the often high toxicity rates associated with a whole-gland approach by reducing cumulative damage to previously irradiated surrounding organs at risk. Conclusion Both biochemical failure and toxicity rates are considerable after salvage radical prostatectomy, Iodine-125 brachytherapy and cryosurgery. Improvements in patient selection using pre-salvage characteristics and other diagnostic modalities can possibly contribute to better outcomes in the future.
  • 46. Chapter1 Chapter 1 44 References 1. Agarwal PK, Sadetsky N, Konety BR, Resnick MI, Carroll PR, Cancer of the Prostate Strategic Urological Research Endeavor (CaPSURE). Treatment failure after primary and salvage therapy for prostate cancer: Likelihood, patterns of care, and outcomes. Cancer. 2008;112(2):307-314. doi: 10.1002/cncr.23161. 2. Heidenreich A, Aus G, Bolla M, et al. EAU guidelines on prostate cancer. Eur Urol. 2008;53(1):68- 80. doi: 10.1016/j.eururo.2007.09.002. 3. Kuban DA, Thames HD, Levy LB, et al. Long-term multi-institutional analysis of stage T1- T2 prostate cancer treated with radiotherapy in the PSA era. Int J Radiat Oncol Biol Phys. 2003;57(4):915-928. 4. Hinnen KA, Battermann JJ, van Roermund JG, et al. Long-term biochemical and survival outcome of 921 patients treated with I-125 permanent prostate brachytherapy. Int J Radiat Oncol Biol Phys. 2010;76(5):1433-1438. doi: 10.1016/j.ijrobp.2009.03.049. 5. Zelefsky MJ, Pei X, Chou JF, et al. Dose escalation for prostate cancer radiotherapy: Predictors of long-term biochemical tumour control and distant metastases-free survival outcomes. Eur Urol. 2011;60(6):1133-1139. doi: 10.1016/j.eururo.2011.08.029 [doi]. 6. Ahmed HU, Pendse D, Illing R, Allen C, van der Meulen JH, Emberton M. Will focal therapy become a standard of care for men with localised prostate cancer? Nat Clin Pract Oncol. 2007;4(11):632-642. doi: 10.1038/ncponc0959. 7. Pound CR, Brawer MK, Partin AW. Evaluation and treatment of men with biochemical prostate- specific antigen recurrence following definitive therapy for clinically localised prostate cancer. Rev Urol. 2001;3(2):72-84. 8. Pucar D, Hricak H, Shukla-Dave A, et al. Clinically significant prostate cancer local recurrence after radiation therapy occurs at the site of primary tumour: Magnetic resonance imaging and step-section pathology evidence. Int J Radiat Oncol Biol Phys. 2007;69(1):62-69. doi: 10.1016/j. ijrobp.2007.03.065. 9. Kimura M, Mouraviev V, Tsivian M, Mayes JM, Satoh T, Polascik TJ. Current salvage methods for recurrent prostate cancer after failure of primary radiotherapy. BJU Int. 2010;105(2):191-201. doi: 10.1111/j.1464-410X.2009.08715.x. 10. Nguyen PL, D'Amico AV, Lee AK, Suh WW. Patient selection, cancer control, and complications after salvage local therapy for postradiation prostate-specific antigen failure: A systematic review of the literature. Cancer. 2007;110(7):1417-1428. doi: 10.1002/cncr.22941. 11. van der Poel HG, Beetsma DB, van Boven H, Horenblas S. Perineal salvage prostatectomy for radiation resistant prostate cancer. Eur Urol. 2007;51(6):1565-71; discussion 1572. doi: 10.1016/j. eururo.2006.08.011. 12. Ash D, Flynn A, Battermann J, et al. ESTRO/EAU/EORTC recommendations on permanent seed implantation for localised prostate cancer. Radiother Oncol. 2000;57(3):315-321.
  • 47. Chapter1 Outcomes of whole-gland salvage in the Netherlands 45 13. Battermann JJ. I-125 implantation for localised prostate cancer: The utrecht university experience. Radiother Oncol. 2000;57(3):269-272. 14. Salembier C, Lavagnini P, Nickers P, et al. Tumour and target volumes in permanent prostate brachytherapy: A supplement to the ESTRO/EAU/EORTC recommendations on prostate brachytherapy. Radiother Oncol. 2007;83(1):3-10. doi: S0167-8140(07)00028-X [pii]. 15. RoachM,3rd,HanksG,ThamesH,Jr,etal.Definingbiochemicalfailurefollowingradiotherapywith or without hormonal therapy in men with clinically localised prostate cancer: Recommendations of the RTOG-ASTRO phoenix consensus conference. Int J Radiat Oncol Biol Phys. 2006;65(4):965- 974. doi: 10.1016/j.ijrobp.2006.04.029. 16. Buyyounouski MK, Hanlon AL, Eisenberg DF, et al. Defining biochemical failure after radiotherapy with and without androgen deprivation for prostate cancer. Int J Radiat Oncol Biol Phys. 2005;63(5):1455-1462. doi: 10.1016/j.ijrobp.2005.05.053. 17. Trotti A, Colevas AD, Setser A, et al. CTCAE v3.0: Development of a comprehensive grading system for the adverse effects of cancer treatment. Semin Radiat Oncol. 2003;13(3):176-181. doi: 10.1016/S1053-4296(03)00031-6. 18. Ward JF, Pagliaro LC, Pisters LL. Salvage therapy for radiorecurrent prostate cancer. Curr Probl Cancer. 2008;32(6):242-271. doi: 10.1016/j.currproblcancer.2008.10.001. 19. Sanderson KM, Penson DF, Cai J, et al. Salvage radical prostatectomy: Quality of life outcomes and long-term oncological control of radiorecurrent prostate cancer. J Urol. 2006;176(5):2025-31; discussion 2031-2. doi: S0022-5347(06)01767-8 [pii]. 20. Moman MR, van der Poel HG, Battermann JJ, Moerland MA, van Vulpen M. Treatment outcome and toxicity after salvage 125-I implantation for prostate cancer recurrences after primary 125-I implantation and external beam radiotherapy. Brachytherapy. 2010;9(2):119-125. doi: 10.1016/j. brachy.2009.06.007. 21. Beyer DC. Permanent brachytherapy as salvage treatment for recurrent prostate cancer. Urology. 1999;54(5):880-883. 22. Ng CK, Moussa M, Downey DB, Chin JL. Salvage cryoablation of the prostate: Followup and analysis of predictive factors for outcome. J Urol. 2007;178(4 Pt 1):1253-7; discussion 1257. doi: 10.1016/j.juro.2007.05.137. 23. Chin JL, Pautler SE, Mouraviev V, Touma N, Moore K, Downey DB. Results of salvage cryoablation of the prostate after radiation: Identifying predictors of treatment failure and complications. J Urol. 2001;165(6 Pt 1):1937-41; discussion 1941-2. 24. Izawa JI, Madsen LT, Scott SM, et al. Salvage cryotherapy for recurrent prostate cancer after radiotherapy: Variables affecting patient outcome. J Clin Oncol. 2002;20(11):2664-2671. 25. Haider MA, Chung P, Sweet J, et al. Dynamic contrast-enhanced magnetic resonance imaging for localisation of recurrent prostate cancer after external beam radiotherapy. Int J Radiat Oncol Biol Phys. 2008;70(2):425-430. doi: 10.1016/j.ijrobp.2007.06.029.
  • 48. Chapter1 Chapter 1 46 26. Barentsz JO, Richenberg J, Clements R, et al. ESUR prostate MR guidelines 2012. Eur Radiol. 2012;22(4):746-757. doi: 10.1007/s00330-011-2377-y; 10.1007/s00330-011-2377-y. 27. Ward JF, Sebo TJ, Blute ML, Zincke H. Salvage surgery for radiorecurrent prostate cancer: Contemporary outcomes. J Urol. 2005;173(4):1156-1160. doi: 10.1097/01.ju.0000155534.54711.60. 28. Bianco FJ,Jr, Scardino PT, Stephenson AJ, Diblasio CJ, Fearn PA, Eastham JA. Long-term oncologic results of salvage radical prostatectomy for locally recurrent prostate cancer after radiotherapy. Int J Radiat Oncol Biol Phys. 2005;62(2):448-453. doi: 10.1016/j.ijrobp.2004.09.049. 29. Grado GL, Collins JM, Kriegshauser JS, et al. Salvage brachytherapy for localised prostate cancer after radiotherapy failure. Urology. 1999;53(1):2-10. 30. PistersLL,RewcastleJC,DonnellyBJ,LugnaniFM,KatzAE,JonesJS.Salvageprostatecryoablation: Initial results from the cryo on-line data registry. J Urol. 2008;180(2):559-63; discussion 563-4. doi: 10.1016/j.juro.2008.04.005. 31. Stephenson AJ, Scardino PT, Bianco FJ,Jr, DiBlasio CJ, Fearn PA, Eastham JA. Morbidity and functional outcomes of salvage radical prostatectomy for locally recurrent prostate cancer after radiation therapy. J Urol. 2004;172(6 Pt 1):2239-2243.  
  • 49.
  • 50. p Daniël A. Smit Duijzentkunst*1 Max Peters*1 Jochem R.N. van der Voort van Zyp1 Marinus A. Moerland1 Marco van Vulpen1 1 Department of Radiation Oncology, University Medical Centre Utrecht, The Netherlands * Both authors contributed equally to this research This chapter requires revisions for: World Journal of Urology
  • 51. Chapter 2 Focal salvage therapy for local prostate cancer recurrences after primary radiotherapy: a comprehensive review
  • 52. Chapter2 Chapter 2 50 Abstract Background/Aim Patients with locally recurrent prostate cancer after primary radiotherapy can be eligible for salvage treatment. Whole-gland salvage techniques carry a high risk of toxicity. A focal salvage approach might reduce the risk of adverse events while maintaining cancer control in carefully selected patients. The aim of this review is to evaluate current literature to assess whether focal salvage leads to a comparable or favourable recurrence rate and less toxicity compared to whole-gland salvage. Methods A literature search was performed using PubMed, Embase and the Cochrane Library. 3015 Articles were screened and assessed for quality. Eight papers (on focal cryoablation (n=3), brachytherapy (n=3) and HIFU (n=2)) were used to report outcomes. Results 1, 2, 3 and 5 year biochemical disease free survival (BDFS) ranges for focal salvage are respectively 69%-100%, 49%-100%, 50%-91% and 46.5%-54.5%. Severe genitourinary, gastrointestinal and sexual function toxicity rates are 0%-33.3%. One study directly compares focal to whole-gland salvage cryotherapy, showing 5-year BDFS of respectively 54.4% and 86.5% with lower toxicity rates for focal salvage patients. Conclusion Provisional data suggest that BDFS rates of focal salvage are in line with those of whole-gland approaches. There is evidence that focal salvage could decrease severe toxicity and preserve erectile function.
  • 53. Focal salvage for radiorecurrent prostate cancer – a review Chapter2 51 Introduction Prostate cancer (PCa) patients primarily treated with external beam radiotherapy (EBRT) or brachytherapy (BT) are at risk of a recurrence, depending on their pre-treatment characteristics. The intermediate and high risk groups can suffer from a biochemical recurrence in over 50% of the cases after 10 years follow-up1,2 . Subsequently, these biochemical recurrences give rise to a risk of developing metastases and (PCa specific) mortality3 . Up to 98% of patients receive (palliative) androgen deprivation therapy (ADT) as treatment after a biochemical recurrence4 . However, a substantial amount of patients harbour organ-confined disease, for which a curative salvage procedure can provide durable disease control5,6 , without exposing patients to the often severe side- effects of ADT7 . Salvage nowadays is usually performed using a whole-gland approach, which is accompanied by a high chance of severe gastro-intestinal (GI), genitourinary (GU) and erectile toxicity due to previous radiation damage to surrounding organs at risk5,6 . Given the evidence from pathology studies that recurrences are frequently localised at the site of the primary largest (index) tumour8,9 , a focal salvage approach, directed solely at the area containing recurrent tumour, might be a viable treatment option for patients with unifocal PCa recurrences without metastatic disease. This way, the serious adverse events associated with whole-gland salvage might be prevented, while cancer control is maintained. The current literature regarding survival and functional outcomes of focal salvage techniques for prostate cancer recurrences after primary radiotherapy is evaluated here. Materials and methods Literature search On 19-08-2015 a systematic literature search was performed in the Pubmed, Embase and Cochrane Library databases. In order to create a sensitive search, the search syntax was build based on domain and determinant (Table 1). After removal of duplicates, 3015 papers were screened, selecting 55 articles with matching domain and determinant. Exclusion criteria are listed in the flowchart (Figure 1). An additional reference search was performed, resulting in 12 studies for this review. Study selection Studies were subjected to a critical appraisal based on an adaptation of the Dutch Cochrane Centre recommendations (ACROBAT-NRSI risk of bias assessment
  • 54. Chapter2 Chapter 2 52 Table 1: Search Syntax     All search terms (title/abstract) for Pubmed, (ab,ti) for Embase, (ti,ab,kw) for Cochrane 1 Salvage 14 Local 2 Therapy 15 Focal 3 Treatment 16 Prostate 4 Rescue 17 Prostatic 5 Cryoablation 18 Cancer 6 Cryosurgery 19 Carcinoma 7 Cryotherapy 20 Adenocarcinoma 8 Ablation 21 Neoplasma 9 Brachytherapy 22 Recurrence 10 HIFU 23 Recurrences 11 "high intensity focused ultrasound" 24 Recurrent 12 Hemi 25 Relapse 13 Partial 26 Radiorecurrent         27 1 or 2 or 3 or 4 or 5 or 6 or 7 or 8 or 9 or 10 or 11 28 12 or 13 or 14 or 15     29 27 and 28     30 16 or 17 #34 Search Results 31 18 or 19 or 20 or 21 Pubmed: 1719 32 22 or 23 or 24 or 25 or 26 Embase: 2811 33 30 and 31 and 32 Cochrane: 199 34 29 and 33 Total: 4729 tool)10 . Studies were graded (+), (+) or (-) on relevance (domain, determinant, outcome) and validity (selection, study population characteristics, exposure, primary outcome, secondary outcomes, follow-up and number of patients) (Table A1 (supplementary file)). All studies were observational cohorts or case series with either prospective or retrospective data acquisition. In that aspect, no study could be considered of high quality. In none of the studies blinding was applied. However, the primary outcome (BDFS) is unlikely to be influenced by information bias, due to the objectivity of the failure definition (Phoenix, i.e. PSA-nadir + 2.0 ng/ml). On the contrary, the secondary outcome is subjective to information bias and is graded in that way. Confounding was not graded, since all studies except one were single-arm
  • 55. Focal salvage for radiorecurrent prostate cancer – a review Chapter2 53 4729 3015 55 11 12 Embase 2811 Pubmed 1719 Removal of duplicates Inclusion criteria Domain = locally recurrent PCa after primary radiotherapy Determinant = focal salvage treatment Exclusion criteria No full text = 6 Review = 10 Conference abstract/ poster = 19 Comment/opinion/ reply = 2 Study protocol = 1 Guideline = 1 Technical report = 2 Reprinted article = 3 Screening title/abstract Full text evaluation Related articles Cochrane 199 Figure 1. Flowchart observational cohorts. The study by De Castro et al.11 describes a two-armed cohort. Here, possible confounding factors are adequately described, but not corrected for in multivariable analysis. Four studies describe case series with 10 patients or less. These were valued as low quality of evidence and therefore excluded. Results Search results A total of 8 studies were eligible for data extraction, describing focal salvage treatment with cryoablation (CA) (n=3), low-dose-rate brachytherapy (LDR- BT) (n=3) and high intensity focused ultrasound (HIFU) (n=2) in a total of 278