You are on page 1of 12

International Journal of Urology (2018) 25, 220--231 doi: 10.1111/iju.

13512

Review Article

Current treatment strategies for advanced prostate cancer


Kazumasa Komura,1 Christopher J Sweeney,2 Teruo Inamoto,1 Naokazu Ibuki,1 Haruhito Azuma1 and
Philip W Kantoff3
1
Department of Urology, Osaka Medical College, Takatsuki, Osaka, Japan, 2Department of Medical Oncology, Dana-Farber Cancer
Institute, Boston, MA, and 3Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA

Abbreviations & Acronyms


Abstract: During the past decade, treatment strategies for patients with advanced
Abi = abiraterone
ADT = androgen deprivation therapy
prostate cancer involving stage IV (T4N0M0, N1M0 or M1) hormone-sensitive prostate
APC = advanced prostate cancer cancer and recurrent prostate cancer after treatment with curative intent, as well as
AR = androgen receptor castration-resistant prostate cancer, have extensively evolved with the introduction and
ASI = androgen signaling inhibitor approval of several new agents including sipuleucel-T, radium-223, abiraterone,
C20 = cabazitaxel 20 mg/m2 enzalutamide and cabazitaxel, all of which have shown significant improvement on
C25 = cabazitaxel 25 mg/m2 overall survival. The appropriate use of these agents and the proper sequencing of these
CRPC = castration-resistant prostate
agents are still not optimized. The results of several recently reported randomized
cancer
CTC = circulating tumor cell
controlled trials and retrospective studies could assist in developing a treatment strategy
ctDNA = circulating tumor DNA for advanced prostate cancer. In addition, prospective studies and molecular
D75 = DNA docetaxel 75 mg/m2 characterization of tumors to address these issues are ongoing.
dNLR = derived neutrophil-to-
lymphocyte ratio Key words: castration-resistant prostate cancer, precision medicine, prostate cancer,
DOC = docetaxel sequential therapy.
Enz = enzalutamide
EZH2 = enhancer of zeste homolog 2
FDA = US Food and Drug
Administration
FISH = fluorescent in situ hybridization Introduction
GCSF = granulocyte colony-
stimulating factor PC is the most common malignancy in men.1 Historically, since Charles Huggins first
HR = hazard ratio reported the effect of ADT for patients with metastatic PC in 1941, suppression of AR signal-
HSPC = hormone-sensitive prostate ing through ADT has remained the mainstay of treatment for metastatic PC for more than
cancer 70 years. Current ADT includes surgical castration or medical castration using luteinizing hor-
IHC = immunohistochemistry mone-releasing hormone agonists or antagonists with or without anti-androgen drugs.
LHRHA = luteinizing hormone-
Although ADT offers near certain remissions lasting 1–2 years in most patients, cancer cells
releasing hormone analog
mCRPC = metastatic castration- become resistant with the emergence of mCRPC. In 2004, docetaxel, a chemotherapeutic inhi-
resistant prostate cancer bitor of microtubule depolymerization, was approved for mCRPC patients; however, docetaxel
NEPC = neuroendocrine prostate offered a modest 2.5 months median prolongation in OS.2,3 Acquired resistance to the drug
cancer eventually emerges.
NLR = neutrophil-to-lymphocyte ratio For the past decade, much progress has been made in the understanding of APC, which
OS = overall survival
includes stage IV (T4N0M0, N1M0 or M1) HSPC and recurrent PC after treatment with cura-
PARP = poly adenosine
diphosphate-ribose polymerase
tive intent as the new high-throughput technologies, such as next-generation sequencing,
PBMC = peripheral blood which has uncovered the mutational landscape of PC. In addition, five additional drugs were
mononuclear cell approved by the FDA for the treatment of CRPC with an improvement of OS, including a
PC = prostate cancer chemotherapeutic (cabazitaxel),4 two ASIs (abiraterone5 and enzalutamide6), an immunothera-
PFS = progression-free survival peutic (sipuleucel-T7) and an alpha-emitting bone-seeking radioisotope (radium-2238). These
PSA = prostate-specific antigen newly developed agents, however, now raise new questions: what constitutes optimal
RB1 = retinoblastoma 1
sequencing or pairing of drugs? And what biomarkers could be used to predict the treatment
RCT = randomized controlled trial
SOC = standard of care
efficacy for precision medicine? By now, a number of prospective and retrospective studies
ZA = zoledronic acid have offered some information to address these issues. In the present article, we review the
overview of the current status for the treatment of APC and provide the latest findings of new
Correspondence: Philip W Kantoff molecular biomarkers for disease prediction.
M.D., Department of Medicine,
Memorial Sloan Kettering Cancer Center,
1275 York Avenue, New York, NY
10065, USA. Email: kantoff@mskcc.org
Five newly developed agents the FDA approved
Received 1 September 2017; accepted
Since docetaxel was approved for mCRPC patients after showing a survival advantage over
9 November 2017. mitoxantrone in 2004, five newly developed drugs were additionally approved by 2014.2,3
Online publication 20 December 2017 Table 1 summarizes phase 3 trials of those drugs.

220 © 2017 The Japanese Urological Association


Current treatment strategy for CRPC

Table 1 Results of randomized clinical trials for advanced prostate cancer drugs

Year Drug name Trial name Patient enrollment Intervention Results (median overall survival)

2004 Docetaxel TAX3273 1006 men with mCRPC Docetaxel 75 mg/m2 every 3 weeks vs Docetaxel every 3 weeks:
docetaxel 30 mg/m2 weekly vs 18.9 months
mitoxantrone 12 mg/m2 every 3 weeks Docetaxel weekly: 17.4 months
(prednisone 5 mg twice daily to all patients) Mitoxantrone: 16.5 months
Docetaxel every 3 weeks vs
mitoxantrone:
HR 0.76, P = 0.009
Docetaxel weekly vs mitoxantrone:
HR 0.91, P = 0.36
2004 Docetaxel SWOG99162 770 men with mCRPC Docetaxel 60 mg/m2 every Docetaxel + estramustine:
3 weeks + estramustine 280 mg 3 times 17.5 months
daily on days 1–5 vs mitoxantrone Mitoxantrone + prednisone:
12 mg/m2 every 3 weeks + prednisone 15.6 months
5 mg twice daily HR 0.80, P = 0.02
2010 Cabazitaxel TROPIC4 755 men who progressed after Cabazitaxel 25 mg/m2 every 3 weeks vs Cabazitaxel: 15.1 months
docetaxel mitoxantrone 12 mg/m2 every 3 weeks Mitoxantrone: 12.7 months
(prednisone 5 mg twice daily to all patients) HR 0.70, P < 0.0001
2010 Sipuleucel-T IMPACT7 512 men with mCRPC without visceral Sipuleucel-T given every 2 weeks 3 times vs Sipuleucel-T: 25.8 months
metastasis† placebo Placebo: 21.7 months
2:1 randomization HR 0.78, P = 0.03
2011 Abiraterone COU-AA-3015 1195 men previously treated with Abiraterone 1000 mg daily vs placebo Abiraterone: 14.8 months
Acetate docetaxel (prednisone 5 mg twice daily to all patients) Placebo: 10.9 months
2:1 randomization HR 0.74, P < 0.001
2012 Enzalutamide AFFIRM6 1199 men previously treated with Enzalutamide 160 mg daily vs placebo Enzalutamide: 18.4 months
docetaxel Placebo: 13.6 months
2:1 randomization HR 0.63, P < 0.001
2012 Abiraterone COU-AA-30213 1088 chemotherapy-na€ıve men with Abiraterone 1000 mg daily vs placebo Abiraterone: 34.7 months
Acetate mCRPC (prednisone 5 mg twice daily to all patients) Placebo: 30.3 months
HR 0.81, P < 0.0033
2013 Radium-223 ALSYMPCA8 921 men with mCRPC with at least Radium-223 every 4 weeks 6 times vs placebo Radium: 14.9 months
two bone metastases, no visceral Placebo: 11.3 months
metastasis, no lymph nodes >3 cm HR 0.70, P < 0.001
2:1 randomization
2014 Enzalutamide PREVAIL19 1717 asymptomatic or minimally Enzalutamide 160 mg vs placebo Enzalutamide: 32.4 months
symptomatic, chemotherapy-na€ıve, Placebo: 30.4 months
no prior abiraterone or HR 0.70, P < 0.01
ketoconazole
2015 Docetaxel CHAARTED22 790 metastatic hormone-sensitive Docetaxel 75 mg/m2 every 3 weeks for 6 ADT + docetaxel: 57.6 months
prostate cancer cycles + ADT vs ADT alone ADT alone: 44.0 months
HR 0.61, P < 0.001
2015 Docetaxel STAMPEDE23 2962 locally advanced high-risk or SOC only vs SOC + ZA (4 mg) every 3 weeks SOC only: 71 months
mCRPC vs SOC + DOC 75 mg/m2 every 3 weeks SOC + ZA: not reached
2:1:1:1 randomization with prednisolone 10 mg daily vs SOC + DOC: 81 months
SOC + ZA + DOC SOC + ZA + DOC: 76 months
SOC vs SOC + ZA
HR 0.94, P = 0.45
SOC vs SOC + DOC
HR 0.78, P = 0.006
SOC vs SOC + ZA + DOC
HR 0.82, P = 0.022
2016 Cabazitaxel FIRSTANA9 1168 patients who had progressed C20 every 3 weeks vs C25 every 3 weeks vs C20: 24.5 months
after castration D75 every 3 weeks (prednisone 5 mg twice C25: 25.2 months
1:1:1 randomization daily to all patients) D75: 24.3 months
C20 vs D75: HR 1.009, P = 0.9967
C25 vs D75: HR 0.97, P = 0.7574
2017 Abiraterone NCT0026847626 1917 locally advanced high-risk or ADT + abiraterone 1000 mg daily vs ADT ADT + abiraterone: 83% of 3 years OS
Acetate metastatic hormone-sensitive alone (prednisone 5 mg twice daily to all ADT alone: 76% of 3 years OS
prostate cancer patients) HR 0.63, P < 0.001
2017 Abiraterone LATITUDE25 1199 metastatic hormone-sensitive ADT + abiraterone 1000 mg daily vs ADT plus ADT + abiraterone: not reached
Acetate prostate cancer placebo (prednisone 5 mg twice daily to all ADT + placebo: 34.7 months
patients) HR 0.62, P < 0.001

†Patients who reported moderate-to-severe PC-related pain and/or use of narcotics for cancer-related pain were also excluded.

© 2017 The Japanese Urological Association 221


K KOMURA ET AL.

the conversion of pregnenolone and progesterone to 17-OH


Sipuleucel-T
pregnenolone and 17-OH progesterone, and from there to
Sipuleucel-T is a cellular immunotherapy consisting of dehydroepiandrosterone and androstenedione, which is the
autologous PBMCs followed by leukapheresis. The penultimate step in testosterone production. Thus, this drug in
patient’s PBMCs are cultured (activated) with a recombi- turns interrupts androgen production at three sources: the tes-
nant human protein (PAP-GM-CSF) consisting of prostatic tis, the adrenal glands and the tumor itself.10–12 In the COU-
acid phosphatase linked to granulocyte-macrophage col- AA-301 trial, 1195 patients previously treated with docetaxel
ony-stimulating factor, and re-injected to the patients three were randomly assigned to abiraterone plus prednisone or
times at 2-week intervals. This therapy was approved in placebo plus prednisone groups.5 The primary end-point of
2010 based on the result from the IMPACT study in OS showed an improvement of OS for 3.9 months compared
which 512 patients having mCRPC without visceral with the placebo. All secondary end-points also favored abi-
metastasis were assigned to a sipuleucel-T arm or a pla- raterone, with a PSA response rate of 29% versus 6%. The
cebo (injection of PBMCs not treated).7 The sipuleucel-T most common side-effect was fatigue, which was similar in
arm had a 4.1-month OS advantage compared with the both groups. There were more mineralocorticoid symptoms
placebo (HR 0.78, 95% CI 0.61–0.98, P = 0.03), whereas consisting of fluid retention in the abiraterone group, and
there was no significant advantage in PFS (3.7 and more hypokalemia in the abiraterone group despite the major-
3.6 months of the median PFS in sipuleucel-T and pla- ity of these side-effects being assigned as grade 1. In the
cebo, respectively) and PSA response rate (2.6% and COU-AA-302 trial, 1088 chemotherapy-na€ıve patients were
1.3% of 50% reduction of PSA in sipuleucel-T and pla- randomized to abiraterone plus prednisone or to a placebo
cebo, respectively). Modest side-effects including chills, plus prednisone.13,14 The primary end-points were OS and
fever and headache were reported, which are comparable radiographic PFS. There was an 8.2-month improvement in
with other new agents. radiographic PFS favoring abiraterone. OS was improved
with abiraterone–prednisone (median not reached, vs
27.2 months for prednisone alone; HR 0.75, 95% CI
Cabazitaxel
0.61–0.93, P = 0.01), but did not cross the efficacy bound-
Cabazitaxel, approved by the FDA in 2010 for patients with ary.13 The final OS results in the trial showed an improve-
mCRPC who progressed after docetaxel,4 is a novel tubulin- ment of OS of 4.4 months in abiraterone group (HR 0.81,
binding taxane that differs from docetaxel because of its 95% CI 0.70–0.93, P = 0.033).14
poor affinity for P-glycoprotein (P-gp), an adenosine triphos-
phate-dependent drug efflux pump. Preclinical and early
Enzalutamide
clinical studies demonstrated that cabazitaxel showed activ-
ity in docetaxel-resistant tumors, and this was confirmed by Enzalutamide is a potent blocker targeting the AR signaling
the TROPIC study. In the study, 755 patients were ran- pathway at several steps, exerting its effect by blocking bind-
domly assigned, and the median OS was 15.1 months in the ing of androgens to AR,15 by inhibiting nuclear translocation
cabazitaxel group and 12.7 months in the mitoxantrone of activated AR16 and by impairing binding of activated AR
group. The HR for death of men treated with cabazitaxel with DNA.17,18 This drug was approved by the FDA in 2012
was 0.70 (95% CI 0.59–0.83, P < 0.0001). The median PFS for mCRPC with progression after docetaxel. In the AFFIRM
was 2.8 months in the cabazitaxel group and 1.4 months in trial, 1199 patients who were previously treated with a doc-
the mitoxantrone group (HR 0.74, 95% CI 0.64–0.86, etaxel chemotherapy were randomly assigned into enzalu-
P < 0.0001). Febrile neutropenia was observed in 8% of tamide or placebo.6 The primary end-point of OS showed an
patients of the cabazitaxel group and 1% in the mitox- improvement of OS for 4.8 months favoring enzalutamide
antrone group. Accordingly, using cabazitaxel for patients (HR 0.63, 95% CI 0.53–0.75, P < 0.001). There was also
with neutrophil counts of <1500 cells/mm3 is a contraindica- improvement in the secondary end-points of radiographic
tion, and the FDA states that prophylactic GCSF should be PFS (median survival of 8.3 months vs 2.9 months; HR 0.40,
considered for patients with high-risk features (age P < 0.001) and the time to the first skeletal-related event
>65 years, poor PS, previous episodes of febrile neutrope- (median survival of 16.7 months vs 13.3 months; HR 0.69,
nia, extensive prior radiation ports, poor nutritional status or P < 0.001). In the PREVAIL trial, 1717 patients with asymp-
other serious comorbidities). The FIRSTANA study recently tomatic or minimally symptomatic mCRPC who had not
showed that 20 mg/m2 was less toxic and as effective as received chemotherapy, abiraterone or ketoconazole were ran-
25 mg/m2 of cabazitaxel, so 20 mg/m2 without GCSF sup- domized to enzalutamide or a placebo.19 The primary end-
port is now the preferred dose.9 points were radiographic PFS and OS. At 12 months, the
radiographic PFS was 65% in the enzalutamide group versus
14% in the placebo group, and median survival had not been
Abiraterone acetate
reached in the enzalutamide group and was 3.9 months in the
Abiraterone, which was first approved by the FDA in 2011 placebo group (HR 0.19, P < 0.001). The median OS were
for use in patients with mCRPC who have been previously 32.4 and 30 months in the enzalutamide arm and the placebo
treated with docetaxel, is a potent P450 c17 (CYP17) inhibi- group, respectively (HR 0.71, 95% CI 0.60–0.84, P < 0.001).
tor, an enzyme that has two distinct activities: 17–20 lyase The most common adverse events in both trials were fatigue,
and 17-alpha hydroxylase. This enzymatic action is used in hot flashes and headache. Of note, in the AFFIRM trial, there

222 © 2017 The Japanese Urological Association


Current treatment strategy for CRPC

were five seizures in the enzalutamide arm and none in the one of the next areas of research was determining whether
placebo arm. Warning of using enzalutamide to the patients combination use or sequential therapy was superior. Further-
with a history of seizures has been described by the FDA, more, the focus was not only on mCRPC. The treatment
which could lead to the result of the PREVAIL trial where strategy for APC including stage IV (T4N0M0, N1M0 or
one seizure was noted in each arm. M1) HSPC and recurrent PC after the treatment with curative
intent are also now evolving.
Radium-223
Radium-223, which was FDA-approved in 2013, is an alpha- Docetaxel as SOC in hormone-sensitive APC
emitting radiopharmaceutical, and selectively targets bone Having shown docetaxel prolonged OS in CRPC, it was then
metastases with alpha particles. Radium is a “calcium evaluated in metastatic hormone-sensitive disease. As shown
mimetic”, which accumulates preferentially in areas of bone in Table 2, in 2013, the result of the GETUG-AFU 15 trial
with increasing turnover, such as osteoblastic metastasis, and was reported in which 385 men with mHSPC were random-
the emitted alpha particle radiation causes double-strand ized to receive ADT plus docetaxel (75 mg/m2 every
breaks in DNA within 100 lm of the range, which offers a 3 weeks, up to nine cycles) or ADT alone.20 Although the
selective effect to the target region without damage to the sur- addition of docetaxel was associated with an improvement in
rounding tissue. In the ALSYMPCA trial, 921 patients were biochemical PFS (22.9 vs 12.9 months; HR 0.7, 95% CI 0.6–
eligible to participate in the study according to the criteria of 0.9, P = 0.0021), there was no improvement in OS with the
mCRPC having two or more bone metastases detected on addition of docetaxel, even with long-term follow up (62.1 vs
skeletal scintigraphy and no known visceral metastases.8 All 48.6 months; HR 0.88, 95% CI 0.68–1.14, P = 0.3).21 In
the patients were receiving the best standard of care, and contrast, the Eastern Cooperative Oncology Group led the
patients who had received docetaxel 4 weeks before the enroll- CHAARTED trial, in which 790 men with mHSPC were ran-
ment were included. The primary end-point was OS, and the domly assigned to ADT or ADT plus six cycles of docetaxel
secondary end-points included time to the first symptomatic chemotherapy given at 75 mg/m2 every 3 weeks and showed
skeletal event and biochemical failure. The final analysis dramatic improvement of OS in the ADT plus docetaxel
showed significant improvement for OS in the radium-223 group than the ADT alone (57.6 vs 44.0 months; HR 0.61,
group compared with the placebo group (14.9 and 95% CI 0.47–0.80, P < 0.001).22 In parallel, the STAM-
11.3 months of median OS in radium-223 and placebo group, PEDE trial, which assigned 2962 men with either high-risk
respectively: HR 0.70, 95% CI 0.58–0.83, P < 0.001). The localized (24%), node-positive (15%) or mHSPC (61%) to
time to the first symptomatic skeletal event, the secondary four treatment arms: SOC alone, SOC plus zoledronic acid,
end-point, were also significantly prolonged in the radium-223 SOC plus docetaxel or SOC plus zoledronic acid and doc-
group compared with the placebo group (median of etaxel, also showed that the addition of docetaxel to SOC eli-
15.6 months vs 9.8 months: HR 0.66, 95% CI 0.52–0.83, cited significant improvement in OS (81.0 vs 71.0 months in
P < 0.001). It should be mentioned that radium-223 was very SOC plus docetaxel vs SOC alone, respectively; HR 0.78,
well tolerated with no increase in grade 3–4 toxicity. The 95% CI 0.66–0.93, P = 0.006).23 It should be mentioned that
FDA approved this drug for mCRPC patients without regard the composition of enrolled patients largely differed in the
to prior docetaxel use, although the design of the ALSYMPCA GETUG-AFU 15 trial compared with the other two larger
trial was intended for patients who were not thought to be eli- studies. CHAARTED was initially designated as a trial for
gible to receive chemotherapy or who chose not to receive it. high-volume metastatic disease, defined as the presence of
visceral metastasis and/or four or more osseous metastases.
The protocol was later amended to allow enrollment of low-
Treatment strategy and biomarkers for
volume disease. The final result was enriched with high-
APC
volume patients (65.8%). In comparison, the GETUG-AFU
As the five new drugs emerged and represented significant 15 trial was not initially powered to evaluate this effect of
improvement in the treatment of mCRPC, it was natural that volume of disease. Subgroup analysis according to volume

Table 2 Median OS in months according to disease volume in RCTs for patients with mHSPC

GETUG-AFU 15 (n = 385)20 CHAARTED (n = 790)22 STAMPEDE: M1 subgroup (n = 1087)23

ADT + ADT + ADT +


Docetaxel ADT HR 95% CI P-value Docetaxel ADT HR 95% CI P-value Docetaxel ADT HR 95% CI P-value

All cohort All cohort All M1 subgroup


60.9 46.5 0.9 0.7–1.2 0.44 57.6 44 0.61 0.47–0.8 <0.001 65 43 0.73 0.59–0.89 0.002
High-volume disease High-volume disease High-volume disease
39.8 35.1 0.78 0.56–1.09 0.14 49.2 32.2 0.6 0.45–0.81 <0.001 - - - - -
Low-volume disease Low-volume disease Low-volume disease
Not reached 83.4 1.02 0.67–1.55 0.9 Not reached Not reached 0.6 0.32–1.13 0.11 - - - - -

© 2017 The Japanese Urological Association 223


K KOMURA ET AL.

disease was later analyzed, and this retrospective evaluation the robust effect of ASIs for the treatment of APC, but also
still did not show significant improvement in OS.21 The med- raise the controversial discussion for the optimal sequence or
ian OS and number of death events were substantially differ- combination use of those agents. The STAMPEDE arm G
ent between these two trials (54.2 months with 176 deaths in cohort involved patients with non-metastatic high-risk disease
GETUG-AFU 15, and 44 months with 237 deaths in (characterized by T3 or T4, tumor-positive lymph nodes;
CHAARTED), implying that the smaller GETUG-15 trial Gleason’s score of 8–10 and/or serum PSA level of ≥40 ng/
coupled with the better prognosis of enrolled patients might mL): 915 and 1002 patients in non-metastatic and metastatic
have resulted in the difference between the trials. A potential groups, respectively. The advantage on OS of combination
selection criteria for benefit in CHAARTED was a prespeci- use of abiraterone was only observed in M1 patients, not in
fied subgroup analysis of patients with high- versus low- M0 patients. The LATITUDE trial enrolled only M1 patients,
volume disease showing an unprecedented 17-month OS and showed a significant improvement on OS by combination
improvement with the addition of docetaxel with high-volume use of abiraterone with ADT. A systematic review of the M1
mHSPC (49.2 vs 32.2 months; HR 0.60, 95% CI 0.45–0.81, cohort in STAMPEDE arm G (1002 patients) and LATI-
P < 0.001), whereas docetaxel originally conferred a rela- TUDE (1199 patients) reported that survival benefit might be
tively modest 2.9-month OS benefit in mCRPC. A long-term greater in younger patients (aged <75 years) despite the small
efficacy in the CHAARTED trial was later reported showing sample size of older patients (137/1002 and 202/1199 in
that this benefit on OS was not seen in patients with low- STAMPEDE arm G and LATITUDE, respectively).27 As
volume disease.24 Nevertheless, the strongly positive sub- these two studies did not define low and high tumor burden
group analysis favoring high-volume disease was sufficiently in their analysis, whether this survival benefit with the combi-
convincing that docetaxel for mHSPC has been embraced by nation of abiraterone is delivered in lower tumor burden
the National Comprehensive Cancer Network, and has since patients remains unknown. Nevertheless, abiraterone should
become a SOC for high-volume of metastatic disease. Given be considered an option in addition to ADT for men with
that subgroup analysis of metastatic patients (n = 1087) in mHSPC.
STAMPEDE trials also showed substantial improvement of
OS for 22 months (65 vs 43 months; HR 0.73, 95% CI 0.59–
Sequential therapy using ASIs
0.89, P = 0.002), we believe that docetaxel should be consid-
ered in addition to ADT in patients with high-volume There have been a number of studies reporting sequential use
mHSPC. of ASIs, namely enzalutamide followed by abiraterone28–32
and abiraterone followed by enzalutamide (Table 3).31–38
Data seem to show a blunted effect of each after the use of
Abiraterone in M1 HSPC patients
the other, suggesting cross-resistance. It remains unclear
How new ASIs, including abiraterone and enzalutamide, which patients benefit from one agent as the first agent com-
should be deployed with other approved agents for the treat- pared with the other. Further investigation is still required in
ment of CRPC has been controversial. As of now, both prospective and randomized settings to validate the optimal
agents are approved as pre-docetaxel and post-docetaxel treat- treatment decision-making. Recently, a phase 4, multicenter,
ment for CRPC.5,6,13,14,19 Recently, two RCTs (LATITUDE25 single-arm, open label study of enzalutamide in 214 patients
and STAMPEDE arm G26) further reported that combination who progressed on abiraterone after treatment of >6 months
use of abiraterone with ADT in mHSPC also offers improve- for CRPC noted a median time to biochemical progression of
ment of OS compared with ADT alone. These data showed 5.7 months and radiographic progression of 8.1 months,

Table 3 Clinical outcomes in the retrospective studies for sequential therapy using abiraterone and enzalutamide

Prior % Patients with PSA decline


Year Authors Sequence docetaxel n of ≥50% by 2nd ASI Median OS
2013 Noonan et al.30 Enz ? Abi + 30 4 11.5 months
2013 Loriot et al.28 Enz ? Abi + 38 8 7.2 months
2017 Terada et al.31 Enz ? Abi 85 13 899 days
2017 Mori et al.32 Enz ? Abi +† 46 6.5 13.5 months
2014 Schrader et al.37 Abi ? Enz + 35 28.6 7.1 months
2014 Badrising et al.34 Abi ? Enz + 61 21 7.3 months
2014 Thomsen et al.38 Abi ? Enz + 24 17 4.8 months
2014 David et al.36 Abi ? Enz + 23 39.1 8.5 months
2015 Brasso et al.35 Abi ? Enz + 137 18 8.3 months
2015 Azad et al.33 Abi ? Enz 47 26 8.6 months
2015 Azad et al.33 Abi ? Enz + 68 22 10.6 months
2017 Terada et al.31 Abi ? Enz + 113 29 919 days
2017 Mori et al.32 Abi ? Enz +† 23 39.1 14 months

†A total of 23 out of 46 in ‘Enz ? Abi’ and nine out of 23 in “Abi ? Enz” treated with prior docetaxel.

224 © 2017 The Japanese Urological Association


Current treatment strategy for CRPC

leading to the conclusion that enzalutamide still remained


Cross-resistance between classes of agents
active in patients with mCRPC previously treated with abi-
raterone.39 It is clear that cross-resistance between abiraterone and enza-
lutamide exists based on the blunted activity of use of one
agent after the other. Shared resistance mechanisms involve
Prognostic factors to assist decision-making
alternative strategies to activate the AR, such as intratumor
The duration of response to prior ADT has been proposed to be steroid biosynthesis,57 activation of the glucocorticoid recep-
the prognostic factor for the efficacy of subsequent ASI,40–42 tor58 and emergence of an AR splice variant, such as
based on several retrospective studies.43–45 These studies ARV7,59 as well as activation of non-AR survival pathways.
consistently showed that shorter response to first-line ADT With regard to the interaction between ASIs and docetaxel,
is associated with poor PSA response rate and shorter PFS. Schweizer et al. reported, in the retrospective study of 119
Those findings were further observed in the post-hoc analy- mCRPC patients, shorter PFS in patients (n = 24) who were
sis from the COU-AA-301 trial, which showed that the treated with abiraterone followed by docetaxel compared with
patients in the lowest quartile of duration of prior ADT had patients (n = 95) treated with only docetaxel (PFS of
the shortest OS and radiographic PFS.46 Another potential 7.6 months in docetaxel and 4.4 months in abiraterone fol-
prognostic factor for the treatment of ASIs is the extent lowed by docetaxel, P = 0.003), implying the cross-resistance
of PSA decline. Brasso et al. first reported that patients between ASIs and docetaxel.60 In contrast, de Bono et al.
who had >30% or 50% of PSA decline with enzalutamide recently reported, in the post-hoc analysis from the COU-
had longer OS compared with patients who had no such AA-302 trial (chemotherapy-na€ıve men with mCRPC), clini-
PSA decline (11.4 vs 7.1 months, P = 0.001, and 12.6 vs cal advantage for subsequent docetaxel treatment in patients
7.4 months, P = 0.007, respectively).35 Subsequently, several with mCRPC whose disease progressed after treatment with
studies reported the clinical value of the PSA decline within abiraterone acetate.61 Although the modest effect might be
4 weeks from the initiation of ASI (Table 4).35,47–50 Impor- observed in patients who were treated with docetaxel after
tantly, all these studies showed that patients who did not progression with an ASI, subsequent chemotherapy still
respond to ASI within 4 weeks had a poor prognosis for not seems to offer benefit for the treatment of CRPC.62 Cabazi-
only PFS, but also OS, implying that other agents, such as taxel, which had benefit for OS in the patients after progres-
taxanes, should be considered for these patients rather than sion with docetaxel (TROPIC), has been consistently reported
trying another ASI. to have less cross-resistance with ASIs in several retrospec-
At the Advanced Prostate Cancer Consensus Conference tive studies.63–66 Biological findings, which showed growth
2017, clinically and biologically relevant issues for the treat- inhibition of enzalutamide-resistant PC cell lines by cabazi-
ment of APC, including prognostic factors, were discussed.51 taxel compared with docetaxel in vivo,67 also support the use
A number of factors potentially predicting the treatment out- of cabazitaxel after ASIs. Given that the FIRSTANA trial
come, such as serum lactate dehydrogenase32,52 and alkaline reported that there was no survival difference when compar-
phosphatase,53 have been recognized as potential markers for ing docetaxel and cabazitaxel as the first chemotherapy to
the treatment of APC. In addition, a number of studies CRPC,9 cabazitaxel should be offered to patients progressing
including post-hoc analysis have further provided new after docetaxel. In addition, the PROSELICA trial showed
insights for a higher NLR predicting lower response rate to non-inferiority for OS of cabazitaxel at a dose of 20 mg/m2
abiraterone,54 and a higher dNLR (dNLR = absolute neu- compared with 25 mg/m2, although the PSA response rate was
trophil count / white blood cells – absolute neutrophil count) higher in 25 mg/m2.68 Of note, in the study, grade 4 neutrope-
predicting shorter OS to docetaxel55 (post-hoc analysis from nia was observed in 21.3% (20 mg/m2) and 48.6% (25 mg/
VENICE and TAX327 studies), whereas cabazitaxel had a m2) of patients, and neutropenic sepsis/infection for 2.2%
comparable effect on OS in patients with a higher NLR in a (20 mg/m2) and 6.1% (25 mg/m2) of patient, suggesting the
post-hoc analysis from TROPIC study.56 Taken together, advantage of 20 mg/m2 with regard to side-effects. These find-
these findings provide some information for clinicians in the ings help with the determination of the optimal dose of cabazi-
decision-making of treatment for APC. taxel for individual patients using cabazitaxel after docetaxel at

Table 4 The impact of PSA decline within 4 weeks from initiation of ASIs

Pre- Post- PSA decline


Year Authors ASI n docetaxel docetaxel (% patients with PSA decline) P-value for OS

2015 Brasso et al.35 Enz 137 0 137 30% decline at 4 weeks (38%) P = 0.001 (11.4 vs 7.1 months)
50% decline at 4 weeks (18%) P = 0.007 (12.6 vs 7.4 months)
2016 Rescigno et al.50 Abi 274 117 157 30% decline at 4 weeks (46%) P < 0.001 (25.8 vs 15.1 months)
2016 Kato et al.49 Enz 51 25 26 30% decline at 4 weeks (60.8%) P = 0.003
50% decline at 4 weeks (45.1%) P = 0.002
2016 Fuerea et al.48 Abi/Enz/Orteronel 118 28 90 30% decline at 4 weeks P = 0.03 (27.9 vs 17.3 months)
50% decline at 4 weeks P < 0.01 (32.2 vs 15.9 months)
2016 Facchini et al.47 Abi 87 0 87 50% decline at 2 weeks (56%) P = 0.01

© 2017 The Japanese Urological Association 225


K KOMURA ET AL.

20 mg/m2 without GCSF support. Finally, the TAXYNERGY detection of TMPRSS2/ERG fusion as a biomarker might
trial recently reported that patients who did not achieve ≥30% have clinical potential for the prediction of taxane sensitiv-
PSA response with taxane after 12 weeks of therapy would ity.88,89
benefit from switching to other taxane, which could serve as a Mutations in the DNA repair pathway are now intensely
prognostic marker for treatment decision-making.69 focused on the field of cancer research. The BRCA2 muta-
tion, for example, is known as a significant predisposition
marker for aggressive prostate cancer,90,91 and defect of
Molecular biomarkers for disease prediction
BRCA2 might be a predictive biomarker for the exceptional
and new therapeutic targets
response to DNA-damaging chemotherapeutics, such as cis-
As shown in Table 5, AR-V7, a truncated splice variant with platin.92 In addition, a phase 2 study (TOPARP-A) showed
no ligand binding domain, has become an attractive biomar- that the oral PARP inhibitor, olaparib, had antitumor activity
ker predicting the effect of ASIs. Investigators have sought to in 33% of mCRPC patients who had progressed after ASIs
develop the methods for the quantitative assessment of this treatment and chemotherapy, with a strikingly higher
splice isoform from CTCs59 and PBMCs.70 The data from response rate in patients with a deleterious mutation of DNA
recent studies suggested that patients harboring AR-V7 in repair genes including BRCA1/2, ATM, Fanconi’s anemia
CTCs have a higher likelihood of primary resistance to ASIs genes and CHEK2 (88%; 14/16 patients) versus patients with-
and might be considered for taxane therapy, implying clinical out those mutations (6%; 2/33 patients).93 Subsequently, a
use as a reliable biomarker for precision medicine.71–73 The phase 3 study was recently carried out to evaluate the effi-
AR-V7 assays, however, require further refinement before cacy and safety of olaparib versus ASIs in mCRPC, which
they are ready for clinical use. Furthermore, ctDNA analyses progressed on the first ASI treatment and has homologous
assessing AR amplification and/or mutation in univariate recombination repair gene mutations (the PROfound trial:
analyses also appear to be prognostic for a poorer response to NCT02987543).
ASIs.74,75 Nakagawa et al. reported that there were some Intratumor heterogeneity renders varied adaptation to the
patients who experienced negative conversion of AR-V7 in targeted therapy. NEPC has been identified as an aggressive
CTCs by treatment with taxanes, suggesting a possibility of phenotype of APC,94,95 and epithelial plasticity in response
re-sensitization from primary resistance to ASIs after taxane to initial ADT reflects developing the clonal evolution to
therapy.76 There are some evidence of the interaction between NEPC from primary adenocarcinoma.96 Inactivated mutations
AR signaling and taxanes, including inhibition of AR nuclear in major tumor suppressors, TP53 and RB1, were shown to
localization by taxanes,69,77,78 AR induced long-non-coding characterize the NEPC or basal-like cell signature, distinct
RNA79 and drug-efflux genes.80 In addition, it is clear that from canonical AR-dependent luminal-like adenocarcinoma,
epigenetic modifications by disease progression affects AR and cells with those mutations were less sensitive to ASIs by
signature leading to tumorgenesis,81 development of CRPC82 decreased AR dependency.96,97 An upregulation of epigenetic
and drug resistance.83 Genetic alternations, such as PTEN regulator, EZH2, was accompanied with the functional loss
loss, have been investigated and proved that patients with this of those tumor suppressors, and its inhibitor was recently
loss seem to be associated with worse outcome.84,85 In addi- reported to restore the sensitivity to ASIs, suggesting an epi-
tion, other genetic alternations cause the dysregulation of AR genetic approach for the treatment of NEPC.98 Of note, the
signaling, which might be exploited to develop a new molec- mutation of TP53 and RB1 was detectable with whole exome
ular biomarker for disease prediction. ERG, one of the ery- sequencing of ctDNA in mCRPC patients, suggesting its
throblast transformation-specific transcription factors, often application to identify this aggressive subtype.99 Given the
fuses with the promoter region of the TMPRSS2 gene numerous genetic alterations potentially serving as a useful
(21q21.2-3), called TMPRSS2/ERG fusion, and upregulation biomarker for precision medicine, an extensive evaluation of
of the ERG expression level by AR binding to the promoter rigorous biomarkers following REMARK guidelines with all
of TMPRSS2 plays a pivotal role in tumorgenesis with co- relevant covariates in the multivariate analysis is required to
operation of other important somatic mutations.86 Additional truly determine the prognostic and predictive value of these
effects of ERG overexpression interacting with microtubule new biomarkers. Furthermore, the assays themselves and
dynamics leading to taxane resistance was shown,87 and appropriate cut-points are currently yet to be optimized.

Table 5 Potential biomarkers predicting the treatment outcome for APC

Target Type of marker Comments


ARv7 CTCs and PBMCs Detection of AR-V7 in CTCs in CRPC patients associated with resistance to ASIs but not
with taxans59,70–73
TMPRSS2-ERG IHC, FISH and ctDNA ERG positive tumor associated with taxane resistance87–89
PTEN IHC and FISH Worse prognosis and low response rate to abiraterone in patients with PTEN loss84,85
BRCA2 IHC, CTCs and FISH Defect of BRCA2 as a predictive biomarker for the exceptional response to DNA damage
chemotherapeutic and PARP inhibitors90–93
TP53, RB1 CTC and ctDNA Mutation of TP53 and RB1 associated with NEPC and higher sensitivity to EZH2 inhibitor94–99

226 © 2017 The Japanese Urological Association


Current treatment strategy for CRPC

be managed according to their individual health status, not


Treatment algorithm for APC to individual
according to age, and has developed a simple assessment of
patients
health status screening for patients aged >70 years, called G8
Evaluating health status in elderly PC patients clearly affects screening (Fig. 1a).100 In short, they proposed a G8 scoring
treatment decision-making. The International Society of Geri- model that classifies patients into three categories according
atric Oncology recommended that older men with PC should to their individual health status; fit, vulnerable and frail.

(a)
G8 geriatric assessment instrument to determine baseline characteristics

Items Score for possible responses G8 screening test


Has food intake declined over the past 3 0 = Severe decrease in food intake
months owing to loss of appetite, digestive
A problems, difficulties with chewing or 1 = Moderate decrase in food intake
swallowing difficulties? 2 = No decrease in food intake
0 = Weight loss of more than 3 kg G8 Score of >14 G8 Score of <14
1 = Does not know
B Weight loss during the past 3 months
2 = Weight loss between 1 kg and
3 kg
0 = Bed or chair bound
1 = Able to get out of bed or chair Reversible impairment Irreversible impairment
C Mobility
but does not go out by by
2 = Goes out geriatric intervention geriatric intervention
0 = Severe dementia or depression
D Neuropsychological problems 1 = Mild dementia
2 = No psychological problems
0 ≤ 19.0 kg/m2
1 = 19.0–20.9 kg/m2
E Body–mass index
2 = 21.0–22.9 kg/m2 Fit Vulnerable Frail
2
3 > 23.0 kg/m
Does the patient take more than three 0 = Yes
F
prescribed drugs per day? 1 = No
Standard treatment Adapted treatment
0 = Not as good
By comparison with other people of the
0.5 = Does not know
G sama age, how does the patient consider
1 = As good Assessment of impairment consists of ADL, IADL, malnutrition, neuropsychological problems, CISR-G.
his health status?
2 = Better ADL: activities of daily living, IADL: instrumental activities of daily living,
0 > 85 years CISR-G: cumulative illness score rating-geriatrics.
H Age 1 = 80–85 years Details in recommendation of the SIOG Prostate Cancer Working Group
2 ≤ 80 years (Droz et al, Lancet Oncol 2014;15:e404-14)
Total score 0–17

(b) mHSPC
or
Recurrent high risk PC after the treatment with curative intent

Low tumor burden High tumor burden

ADT ADT + abiraterone ADT + docetaxel


(M0 or M1) (M1) (M1)

ADT response <12 months G8 Fit


No Yes Frail or
Visceral metastasis screening vulnerable
Symptomatic

ASIs Docetaxel Enzalutamide Docetaxel ASIs


PSA decline <30% at 4 weeks

Fit Fit Fit G8 Fit


Frail G8 or Frail G8 or Frail G8 or Frail or
screening screening screening screening vulnerable
vulnerable vulnerable vulnerable

ASIs Docetaxel ASIs Cabazitaxel Enzalutamide Cabazitaxel ASIs Cabazitaxel

G8 Fit
Frail or
screening vulnerable

ASIs Cabazitaxel ASIs Enzalutamide ASIs

* Radium-223 is considered for mCRPC with symptomatic bone


and non-visceral metastases.
ASIs

Fig. 1 (a) Left panel: the G8 screening questionnaire. Right panel: decision tree to determine patient health status. (b) Putative algorithm for the treatment of
APC based on the provided findings.

© 2017 The Japanese Urological Association 227


K KOMURA ET AL.

Patients assigned to “fit” should undergo the standard treat- an appropriate option for patients with symptomatic bone and
ment option as younger patients. “Vulnerable” patients who non-visceral metastases. Comparable toxicity when using
have reversible impairment by adequate medical intervention radium-223 with other ASIs was reported,104 and a prelimi-
should also receive standard treatment. “Frail” patients with nary report from a phase 1/2a study showed that radium-223
non-reversible impairment should be considered for adapted with docetaxel had greater percentage decline in total alkaline
treatment. An international collaborative study subsequently phosphatase and in bone formation markers, bone alkaline
showed an advantage of using G8 score to assess tolerability phosphatase and P1NP, than the treatment with only doc-
for the treatment with taxanes, and concluded that treatment etaxel.105 Phase 3 clinical trials are ongoing assessing the
decision-making should be determined by individual health efficacy of radium-223 with abiraterone or enzalutamide to
assessment, not by age.101 determine whether the combination is required.
Figure 1b shows the putative algorithm for the treatment In conclusion, accumulative evidence from RCTs showed
of APC based on the provided findings. A result of a system- that incorporation of docetaxel as a SOC for high metastatic
atic review, which analyzed 13 retrospective studies of burden HSPC offers a benefit in OS, and abiraterone will be
sequential therapy for patients after progression on docetaxel, considered as another option with the results of recent
showed that a sequence that includes cabazitaxel seems to phase 3 studies. Ultimately, access to the agents will be the
confer an advantage for OS compared with successive use of major determining factor. In addition, available retrospective
ASIs, indicating that the standard option after docetaxel studies potentially revealed predictive value including dura-
includes subsequent cabazitaxel for patients assigned to “fit tion of ADT response and the extent of PSA decline by ASIs,
or vulnerable” health status.102 It should be mentioned that to assist in treatment decision-making and deciding which
ASIs might have some effect after progression on cabazitaxel, agent to use next. A number of RCTs are currently ongoing
and patients whose health status descends to “frail” as a (Table 6) to further answer several clinical questions for
result of progression of the disease could still benefit from sequential and combination therapy. Of them, ENZAMET
subsequent treatment with ASIs.103 Radium-223 is considered (enzalutamide with ADT in mHSPC), NCT01650194

Table 6 Active clinical trials to determine optimal sequence or combination using new drugs for APC

Clinical trial Study


identification Trial name start Phase Patients Intervention Primary outcome

NCT01650194 2012 II mCRPC Enzalutamide + abiratetone + prednisone vs PSA response and safety
enzalutamide or abiraterone or prednisone
NCT01995513 PLATO 2013 IV Chemo-na€ıve Enzalutamide + abiratetone + prednisone vs PFS
mCRPC placebo + abiratetone + prednisone
NCT01957436 PEACE-1 2013 III mHSPC ADT  docetaxel vs ADT + abiraterone  OS and PFS
docetaxel vs radiation + ADT  docetaxel +
radiation vs radiation + ADT + abiraterone
NCT01949337 ALLIANCE 2013 III mCRPC Enzalutamide vs enzalutamide + abiratetone + OS
prednisone
NCT02125357 2014 II mCRPC Abiraterone followed by enzalutamide vs PSA response rate to 2nd ASI
enzalutamide followed by abiraterone
NCT02200614 ARAMIS 2014 III High-risk non-metastatic ODM-201 vs placebo Metastatic free survival
CRPC
NCT02429193 BARRIER-P 2015 II mCRPC Enzalutamide + abiratetone + prednisone vs Change in AR abnormalities
enzalutamide or abiraterone or prednisone (ARv7 or AR mutation)
NCT02485691 CARD 2015 III mCRPC after progression Cabazitaxel + prednisone vs abiraterone or rPFS
on docetaxel and first enzalutamide + prednisone
ASI
NCT02489318 TITAN 2015 III mHSPC ARN-509 + ADT  docetaxel vs placebo OS and rPFS
+ ADT  docetaxel
NCT02446405 ENZAMET 2015 III mHSPC Enzalutamide + LHRHA or surgical castration vs OS
non-steroidal anti-androgen vs LHRHA or
surgical castration
NCT02799602 ARASENS 2016 III mHSPC ODM-201 + ADT + docetaxel vs OS
placebo + ADT + docetaxel
NCT02677896 ARCHES 2016 III mHSPC Enzaltamide + ADT vs placebo + ADT rPFS
NCT02987543 PROfound 2016 III mCRPC with homologous Olaparib vs enzalutamide or abiraterone rPFS
recombination repair
deficiency
NCT02446444 ENZARAD 2017 III High-risk localized PC Enzaltamide + LHRHA + radiation vs non- OS
steroidal anti-androgen + LHRHA + radiation

228 © 2017 The Japanese Urological Association


Current treatment strategy for CRPC

(combination of ASIs in mCRPC) and NCT02125357 (cross- 15 Chen Y, Clegg NJ, Scher HI. Anti-androgens and androgen-depleting thera-
pies in prostate cancer: new agents for an established target. Lancet Oncol.
over with ASIs in mCRPC) have recently completed accrual,
2009; 10: 981–91.
and will provide new evidence for the treatment of APC. 16 Guerrero J, Alfaro IE, Gomez F, Protter AA, Bernales S. Enzalutamide, an
Nevertheless, much work including biomarkers development androgen receptor signaling inhibitor, induces tumor regression in a mouse
is required to clarify that best treatment strategy for individ- model of castration-resistant prostate cancer. Prostate 2013; 73: 1291–305.
ual patients. 17 Dalal K, Roshan-Moniri M, Sharma A et al. Selectively targeting the DNA-
binding domain of the androgen receptor as a prospective therapy for pros-
tate cancer. J. Biol. Chem. 2014; 292: 4359.
Conflict of interest 18 Hoffman-Censits J, Kelly WK. Enzalutamide: a novel antiandrogen for
patients with castrate-resistant prostate cancer. Clin. Cancer Res. 2013; 19:
PWK is a scientific advisory or data safety monitoring board 1335–9.
member for Astellas, Bayer, Bellicum, BIND Biosciences, 19 Beer TM, Armstrong AJ, Rathkopf DE et al. Enzalutamide in metastatic
prostate cancer before chemotherapy. N. Engl. J. Med. 2014; 371: 424–
BN ImmunoTherapeutics, DRGT, Genetech/Roche, Ipsen
33.
Pharmaceuticals, Janssen, Metamark (no longer in business), 20 Gravis G, Fizazi K, Joly F et al. Androgen-deprivation therapy alone or
Merck, MTG Therapeutics, New England Research Institutes, with docetaxel in non-castrate metastatic prostate cancer (GETUG-AFU 15):
Omnitura, OncoCellMDX, OncoGenex, Progeniry, Sanofi, a randomised, open-label, phase 3 trial. Lancet Oncol. 2013; 14: 149–58.
Tarveda Therapeutics and Thermo Fisher, and has investment 21 Gravis G, Boher JM, Joly F et al. Androgen deprivation therapy (ADT) plus
docetaxel versus ADT alone in metastatic non castrate prostate cancer:
interest with Bellicum, DRGT and Tarveda Therapeutics. CJS impact of metastatic burden and long-term survival analysis of the random-
has received consulting fees from Astellas, Bayer, Genetech, ized phase 3 GETUG-AFU15 trial. Eur. Urol. 2016; 70: 256–62.
Janssen, Pfizer and Sanofi, and has received research funding 22 Sweeney CJ, Chen YH, Carducci M et al. Chemohormonal therapy in meta-
from Astellas, Janssen, Sotio and Sanofi. static hormone-sensitive prostate cancer. N. Engl. J. Med. 2015; 373: 737–
46.
23 James ND, Sydes MR, Clarke NW et al. Addition of docetaxel, zoledronic
References acid, or both to first-line long-term hormone therapy in prostate cancer
(STAMPEDE): survival results from an adaptive, multiarm, multistage, plat-
1 Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J. Clin. form randomised controlled trial. Lancet 2016; 387: 1163–77.
2017; 67: 7–30. 24 Sweeney C, Chen YH, Liu G et al. Long term efficacy and QOL data of
2 Petrylak DP, Tangen CM, Hussain MH et al. Docetaxel and estramustine chemohormonal therapy (C-HT) in low and high volume hormone na€ıve
compared with mitoxantrone and prednisone for advanced refractory pros- metastatic prostate cancer (PrCa): E3805 CHAARTED trial. Ann. Oncol.
tate cancer. N. Engl. J. Med. 2004; 351: 1513–20. 2016; 27: 720PD-PD.
3 Tannock IF, de Wit R, Berry WR et al. Docetaxel plus prednisone or 25 Fizazi K, Tran N, Fein L et al. Abiraterone plus prednisone in metastatic,
mitoxantrone plus prednisone for advanced prostate cancer. N. Engl. J. castration-sensitive prostate cancer. N. Engl. J. Med. 2017; 377: 352–60.
Med. 2004; 351: 1502–12. 26 James ND, de Bono JS, Spears MR et al. Abiraterone for prostate cancer
4 de Bono JS, Oudard S, Ozguroglu M et al. Prednisone plus cabazitaxel or not previously treated with hormone therapy. N. Engl. J. Med. 2017; 377:
mitoxantrone for metastatic castration-resistant prostate cancer progressing 338–51.
after docetaxel treatment: a randomised open-label trial. Lancet 2010; 376: 27 Rydzewska LHM, Burdett S, Vale CL et al. Adding abiraterone to androgen
1147–54. deprivation therapy in men with metastatic hormone-sensitive prostate can-
5 de Bono JS, Logothetis CJ, Molina A et al. Abiraterone and increased sur- cer: a systematic review and meta-analysis. Eur. J. Cancer 2017; 84: 88–
vival in metastatic prostate cancer. N. Engl. J. Med. 2011; 364: 1995–2005. 101.
6 Scher HI, Fizazi K, Saad F et al. Increased survival with enzalutamide 28 Loriot Y, Bianchini D, Ileana E et al. Antitumour activity of abiraterone
in prostate cancer after chemotherapy. N. Engl. J. Med. 2012; 367: acetate against metastatic castration-resistant prostate cancer progressing
1187–97. after docetaxel and enzalutamide (MDV3100). Ann. Oncol. 2013; 24: 1807–
7 Kantoff PW, Higano CS, Shore ND et al. Sipuleucel-T immunotherapy for 12.
castration-resistant prostate cancer. N. Engl. J. Med. 2010; 363: 411–22. 29 Maughan BL, Luber B, Nadal R, Antonarakis ES. Comparing sequencing of
8 Parker C, Nilsson S, Heinrich D et al. Alpha emitter radium-223 and sur- abiraterone and enzalutamide in men with metastatic castration-resistant
vival in metastatic prostate cancer. N. Engl. J. Med. 2013; 369: 213–23. prostate cancer: a retrospective study. Prostate 2017; 77: 33–40.
9 Sartor AO, Stephane O, Lisa S et al. Cabazitaxel vs docetaxel in chemother- 30 Noonan KL, North S, Bitting RL, Armstrong AJ, Ellard SL, Chi KN. Clini-
apy-naive (CN) patients with metastatic castration-resistant prostate cancer cal activity of abiraterone acetate in patients with metastatic castration-resis-
(mCRPC): a three-arm phase III study (FIRSTANA). J. Clin. Oncol. 2016; tant prostate cancer progressing after enzalutamide. Ann. Oncol. 2013; 24:
34: 5006. 1802–7.
10 Attard G, Belldegrun AS, de Bono JS. Selective blockade of androgenic 31 Terada N, Maughan BL, Akamatsu S et al. Exploring the optimal sequence
steroid synthesis by novel lyase inhibitors as a therapeutic strategy for treat- of abiraterone and enzalutamide in patients with chemotherapy-naive castra-
ing metastatic prostate cancer. BJU Int. 2005; 96: 1241–6. tion-resistant prostate cancer: the Kyoto-Baltimore collaboration. Int. J.
11 Barrie SE, Haynes BP, Potter GA et al. Biochemistry and pharmacokinetics Urol. 2017; 24: 441–8.
of potent non-steroidal cytochrome P450(17alpha) inhibitors. J. Steroid Bio- 32 Mori K, Kimura T, Onuma H et al. Lactate dehydrogenase predicts com-
chem. Mol. Biol. 1997; 60: 347–51. bined progression-free survival after sequential therapy with abiraterone and
12 Potter GA, Barrie SE, Jarman M, Rowlands MG. Novel steroidal inhibitors enzalutamide for patients with castration-resistant prostate cancer. Prostate
of human cytochrome P45017 alpha (17 alpha-hydroxylase-C17,20-lyase): 2017; 77: 1144–50.
potential agents for the treatment of prostatic cancer. J. Med. Chem. 1995; 33 Azad AA, Eigl BJ, Murray RN, Kollmannsberger C, Chi KN. Efficacy
38: 2463–71. of enzalutamide following abiraterone acetate in chemotherapy-naive
13 Ryan CJ, Smith MR, de Bono JS et al. Abiraterone in metastatic prostate metastatic castration-resistant prostate cancer patients. Eur. Urol. 2015a;
cancer without previous chemotherapy. N. Engl. J. Med. 2013; 368: 138– 67: 23–9.
48. 34 Badrising S, van der Noort V, van Oort IM et al. Clinical activity and toler-
14 Ryan CJ, Smith MR, Fizazi K et al. Abiraterone acetate plus prednisone ability of enzalutamide (MDV3100) in patients with metastatic, castration-
versus placebo plus prednisone in chemotherapy-naive men with metastatic resistant prostate cancer who progress after docetaxel and abiraterone treat-
castration-resistant prostate cancer (COU-AA-302): final overall survival ment. Cancer 2014; 120: 968–75.
analysis of a randomised, double-blind, placebo-controlled phase 3 study. 35 Brasso K, Thomsen FB, Schrader AJ et al. Enzalutamide antitumour activity
Lancet Oncol. 2015; 16: 152–60. against metastatic castration-resistant prostate cancer previously treated with

© 2017 The Japanese Urological Association 229


K KOMURA ET AL.

docetaxel and abiraterone: a multicentre analysis. Eur. Urol. 2015; 68: 317– second-line chemotherapy for advanced prostate cancer independent of base-
24. line steroid use. Ann. Oncol. 2015; 26: 750–5.
36 David T, Natalie C, Omi P. Enzalutamide after failure of docetaxel and abi- 57 Chang KH, Li R, Kuri B et al. A gain-of-function mutation in DHT synthe-
raterone in metastatic castrate resistant prostate cancer (mCRPC): results sis in castration-resistant prostate cancer. Cell 2013; 154: 1074–84.
from an expanded access program. J. Clin. Oncol. 2014; 32: 188. 58 Arora VK, Schenkein E, Murali R et al. Glucocorticoid receptor confers
37 Schrader AJ, Boegemann M, Ohlmann CH et al. Enzalutamide in castra- resistance to antiandrogens by bypassing androgen receptor blockade. Cell
tion-resistant prostate cancer patients progressing after docetaxel and abi- 2013; 155: 1309–22.
raterone. Eur. Urol. 2014; 65: 30–6. 59 Antonarakis ES, Lu C, Wang H et al. AR-V7 and resistance to enzalu-
38 Thomsen FB, Roder MA, Rathenborg P, Brasso K, Borre M, Iversen P. tamide and abiraterone in prostate cancer. N. Engl. J. Med. 2014; 371:
Enzalutamide treatment in patients with metastatic castration-resistant pros- 1028–38.
tate cancer progressing after chemotherapy and abiraterone acetate. Scand. 60 Schweizer MT, Zhou XC, Wang H et al. The influence of prior abiraterone
J. Urol. 2014; 48: 268–75. treatment on the clinical activity of docetaxel in men with metastatic castra-
39 De Bono JS, Chowdhury S, Feyerabend S et al. Efficacy and safety of enza- tion-resistant prostate cancer. Eur. Urol. 2014; 66: 646–52.
lutamide (ENZA) in patients with metastatic castration-resistant prostate can- 61 de Bono JS, Smith MR, Saad F et al. Subsequent chemotherapy and treat-
cer (mCRPC) previously treated with abiraterone acetate (Abi): a ment patterns after abiraterone acetate in patients with metastatic castration-
multicenter, single-arm, open-label study. J. Clin. Oncol. 2017a; 35: 165. resistant prostate cancer: post hoc analysis of COU-AA-302. Eur. Urol.
40 Sonpavde G, Wang CG, Galsky MD, Oh WK, Armstrong AJ. Cytotoxic 2017b; 71: 656–64.
chemotherapy in the contemporary management of metastatic castration- 62 Ueda Y, Matsubara N, Takizawa I et al. A multicenter retrospective analysis
resistant prostate cancer (mCRPC). BJU Int. 2015; 116: 17–29. of sequential treatment of abiraterone acetate followed by docetaxel in Japa-
41 Chi K, Hotte SJ, Joshua AM et al. Treatment of mCRPC in the AR-axis-tar- nese patients with metastatic castration-resistant prostate cancer. Jpn. J. Clin.
geted therapy-resistant state. Ann. Oncol. 2015; 26: 2044–56. Oncol. 2015; 45: 774–9.
42 Fitzpatrick JM, Bellmunt J, Fizazi K et al. Optimal management of meta- 63 van Soest RJ, Nieuweboer AJM, de Morree ES et al. The influence of prior
static castration-resistant prostate cancer: highlights from a European Expert novel androgen receptor targeted therapy on the efficacy of cabazitaxel in
Consensus Panel. Eur. J. Cancer 2014; 50: 1617–27. men with metastatic castration-resistant prostate cancer. Eur. J. Cancer
43 Yohann L, Christophe M, Laurence A et al. Personalizing treatment in 2015b; 51: 2562–9.
patients with castrate-resistant prostate cancer: a study of predictive factors 64 Sella A, Sella T, Peer A et al. Activity of cabazitaxel after docetaxel and
for secondary endocrine therapies activity. J. Clin. Oncol. 2012; 30: 213. abiraterone acetate therapy in patients with castration-resistant prostate can-
44 Loriot Y, Eymard JC, Patrikidou A et al. Prior long response to androgen cer. Clin. Genitourin. Cancer 2014; 12: 428–32.
deprivation predicts response to next-generation androgen receptor axis tar- 65 Pezaro CJ, Omlin AG, Altavilla A et al. Activity of cabazitaxel in castra-
geted drugs in castration resistant prostate cancer. Eur. J. Cancer 2015; 51: tion-resistant prostate cancer progressing after docetaxel and next-generation
1946–52. endocrine agents. Eur. Urol. 2014; 66: 459–65.
45 Antoine A, Denis M, Aude F et al. Duration of response to androgen-depri- 66 Al Nakouzi N, Le Moulec S, Albiges L et al. Cabazitaxel remains active in
vation therapy (ADT) and efficacy of secondary hormone therapy, docetaxel patients progressing after docetaxel followed by novel androgen receptor
(D), and cabazitaxel (C) in metastatic castration-resistant prostate cancer pathway targeted therapies. Eur. Urol. 2015; 68: 228–35.
(mCRPC). J. Clin. Oncol. 2014; 32: 282. 67 van Soest RJ, de Morree ES, Kweldam CF et al. Targeting the androgen
46 Bellmunt J, Kheoh T, Yu MK et al. Prior endocrine therapy impact on abi- receptor confers in vivo cross-resistance between enzalutamide and doc-
raterone acetate clinical efficacy in metastatic castration-resistant prostate etaxel, but not cabazitaxel, in castration-resistant prostate cancer. Eur. Urol.
cancer: post-hoc analysis of randomised phase 3 studies. Eur. Urol. 2016; 2015c; 67: 981–5.
69: 924–32. 68 De Bono JS, Hardy-Bessard A-C, Kim C-S et al. Phase III non-inferiority
47 Facchini G, Caffo O, Ortega C et al. Very early PSA response to abi- study of cabazitaxel (C) 20 mg/m2 (C20) versus 25 mg/m2 (C25) in patients
raterone in mCRPC patients: a novel prognostic factor predicting overall (pts) with metastatic castration-resistant prostate cancer (mCRPC) previously
survival. Front. Pharmacol. 2016; 7: 123. treated with docetaxel (D). J. Clin. Oncol. 2016; 34: 5008.
48 Fuerea A, Baciarello G, Patrikidou A et al. Early PSA response is an inde- 69 Antonarakis ES, Tagawa ST, Galletti G et al. Randomized, noncompara-
pendent prognostic factor in patients with metastatic castration-resistant tive, phase II trial of early switch from docetaxel to cabazitaxel or vice
prostate cancer treated with next-generation androgen pathway inhibitors. versa, with integrated biomarker analysis, in men with chemotherapy-
Eur. J. Cancer 2016; 61: 44–51. naive, metastatic, castration-resistant prostate cancer. J. Clin. Oncol.
49 Kato H, Furuya Y, Miyazawa Y et al. Consequences of an early PSA 2017; 35: 3181–8.
response to enzalutamide treatment for Japanese patients with metastatic cas- 70 Qu F, Xie W, Nakabayashi M et al. Association of AR-V7 and prostate-
tration-resistant prostate cancer. Anticancer Res. 2016; 36: 6141–9. specific antigen RNA levels in blood with efficacy of abiraterone acetate
50 Rescigno P, Lorente D, Bianchini D et al. Prostate-specific antigen decline and enzalutamide treatment in men with prostate cancer. Clin. Cancer Res.
after 4 weeks of treatment with abiraterone acetate and overall survival in 2017; 23: 726–34.
patients with metastatic castration-resistant prostate cancer. Eur. Urol. 2016; 71 Antonarakis ES, Lu C, Luber B et al. Androgen receptor splice variant 7
70: 724–31. and efficacy of taxane chemotherapy in patients with metastatic castration-
51 Gillessen S, Attard G, Beer TM et al. Management of patients with resistant prostate cancer. JAMA Oncol. 2015; 1: 582–91.
advanced prostate cancer: the report of the advanced prostate cancer consen- 72 Scher HI, Lu D, Schreiber NA et al. Association of ar-v7 on circulating
sus conference APCCC 2017. Eur. Urol. 2017; 17: 30497–9. tumor cells as a treatment-specific biomarker with outcomes and survival in
52 Wulaningsih W, Holmberg L, Garmo H et al. Serum lactate dehydrogenase castration-resistant prostate cancer. JAMA Oncol. 2016; 2: 1441–9.
and survival following cancer diagnosis. Br. J. Cancer 2015; 113: 1389–96. 73 Onstenk W, Sieuwerts AM, Kraan J et al. Efficacy of cabazitaxel in castra-
53 Gravis G, Boher JM, Fizazi K et al. Prognostic factors for survival in non- tion-resistant prostate cancer is independent of the presence of AR-V7 in
castrate metastatic prostate cancer: validation of the glass model and devel- circulating tumor cells. Eur. Urol. 2015; 68: 939–45.
opment of a novel simplified prognostic model. Eur. Urol. 2015; 68: 196– 74 Azad AA, Volik SV, Wyatt AW et al. Androgen receptor gene aberra-
204. tions in circulating cell-free DNA: biomarkers of therapeutic resistance in
54 Leibowitz-Amit R, Templeton AJ, Omlin A et al. Clinical variables associ- castration-resistant prostate cancer. Clin. Cancer Res. 2015b; 21: 2315–
ated with PSA response to abiraterone acetate in patients with metastatic 24.
castration-resistant prostate cancer. Ann. Oncol. 2014; 25: 657–62. 75 Romanel A, Gasi Tandefelt D, Conteduca V et al. Plasma AR and abi-
55 van Soest RJ, Templeton AJ, Vera-Badillo FE et al. Neutrophil-to-lympho- raterone-resistant prostate cancer. Sci. Transl. Med. 2015; 7: 312re10.
cyte ratio as a prognostic biomarker for men with metastatic castration-resis- 76 Nakazawa M, Lu C, Chen Y et al. Serial blood-based analysis of AR-V7 in
tant prostate cancer receiving first-line chemotherapy: data from two men with advanced prostate cancer. Ann. Oncol. 2015; 26: 1859–65.
randomized phase III trials. Ann. Oncol. 2015a; 26: 743–9. 77 Zhu ML, Horbinski CM, Garzotto M, Qian DZ, Beer TM, Kyprianou N.
56 Lorente D, Mateo J, Templeton AJ et al. Baseline neutrophil-lymphocyte Tubulin-targeting chemotherapy impairs androgen receptor activity in pros-
ratio (NLR) is associated with survival and response to treatment with tate cancer. Cancer Res. 2010; 70: 7992–8002.

230 © 2017 The Japanese Urological Association


Current treatment strategy for CRPC

78 Martin SK, Pu H, Penticuff JC, Cao Z, Horbinski C, Kyprianou N. Multinu- 93 Mateo J, Carreira S, Sandhu S et al. DNA-repair defects and olaparib in
cleation and mesenchymal-to-epithelial transition alleviate resistance to com- metastatic prostate cancer. N. Engl. J. Med. 2015; 373: 1697–708.
bined cabazitaxel and antiandrogen therapy in advanced prostate cancer. 94 Beltran H, Rickman DS, Park K et al. Molecular characterization of neu-
Cancer Res. 2016; 76: 912–26. roendocrine prostate cancer and identification of new drug targets. Cancer
79 Misawa A, Takayama K, Urano T, Inoue S. Androgen-induced long non- Discov. 2011; 1: 487–95.
coding RNA (lncRNA) SOCS2-AS1 promotes cell growth and inhibits 95 Epstein JI, Amin MB, Beltran H et al. Proposed morphologic classification
apoptosis in prostate cancer cells. J. Biol. Chem. 2016; 291: 17861–80. of prostate cancer with neuroendocrine differentiation. Am. J. Surg. Pathol.
80 Zhu Y, Liu C, Armstrong C, Lou W, Sandher A, Gao AC. Antiandrogens 2014; 38: 756–67.
inhibit ABCB1 efflux and ATpase activity and reverse docetaxel resistance 96 Beltran H, Prandi D, Mosquera JM et al. Divergent clonal evolution of cas-
in advanced prostate cancer. Clin. Cancer Res. 2015; 21: 4133–42. tration-resistant neuroendocrine prostate cancer. Nat. Med. 2016; 22: 298–
81 Pomerantz MM, Li F, Takeda DY et al. The androgen receptor cistrome is 305.
extensively reprogrammed in human prostate tumorigenesis. Nat. Genet. 97 Mu P, Zhang Z, Benelli M et al. SOX2 promotes lineage plasticity and
2015; 47: 1346–51. antiandrogen resistance in TP53- and RB1-deficient prostate cancer. Science
82 Wang Q, Li W, Zhang Y et al. Androgen receptor regulates a distinct transcrip- 2017; 355: 84–8.
tion program in androgen-independent prostate cancer. Cell 2009; 138: 245–56. 98 Ku SY, Rosario S, Wang Y et al. Rb1 and Trp53 cooperate to suppress
83 Komura K, Jeong SH, Hinohara K et al. Resistance to docetaxel in prostate prostate cancer lineage plasticity, metastasis, and antiandrogen resistance.
cancer is associated with androgen receptor activation and loss of KDM5D Science 2017; 355: 78–83.
expression. Proc. Natl Acad. Sci. USA 2016; 113: 6259–64. 99 Beltran H, Romanel A, Casiraghi N et al. Whole exome sequencing (WES)
84 Punnoose EA, Ferraldeschi R, Szafer-Glusman E et al. PTEN loss in circulat- of circulating tumor DNA (ctDNA) in patients with neuroendocrine prostate
ing tumour cells correlates with PTEN loss in fresh tumour tissue from castra- cancer (NEPC) informs tumor heterogeneity. J. Clin. Oncol. 2017; 35:
tion-resistant prostate cancer patients. Br. J. Cancer 2015; 113: 1225–33. 5011.
85 Ferraldeschi R, Nava Rodrigues D, Riisnaes R et al. PTEN protein loss and 100 Droz J-P, Aapro M, Balducci L et al. Management of prostate cancer in
clinical outcome from castration-resistant prostate cancer treated with abi- older patients: updated recommendations of a working group of the Inter-
raterone acetate. Eur. Urol. 2015; 67: 795–802. national Society of Geriatric Oncology. Lancet Oncol. 2014; 15: e404–
86 Tomlins SA, Rhodes DR, Perner S et al. Recurrent fusion of TMPRSS2 and 14.
ETS transcription factor genes in prostate cancer. Science 2005; 310: 644–8. 101 Droz J-P, Efstathiou E, Yildirim A et al. First-line treatment in senior
87 Galletti G, Matov A, Beltran H et al. ERG induces taxane resistance in cas- adults with metastatic castration-resistant prostate cancer: a prospective
tration-resistant prostate cancer. Nat. Commun. 2014; 5: 5548. international registry. Urol. Oncol. Sem. Original Invest. 2016; 34:
88 Rajpar S, Carmel A, Merabet Z et al. The benefit of combining docetaxel to 234.e21–e29.
androgen deprivation therapy in localized and metastatic castration-sensitive 102 Maines F, Caffo O, Veccia A et al. Sequencing new agents after docetaxel
prostate cancer as predicted by ERG status: An analysis of two GETUG in patients with metastatic castration-resistant prostate cancer. Crit. Rev.
phase III trials. J. Clin. Oncol. 2017; 35: 5012. Oncol. Hematol. 2015; 96: 498–506.
89 Reig O, Marin-Aguilera M, Carrera G et al. TMPRSS2-ERG in blood and 103 Caffo O, De Giorgi U, Fratino L et al. Clinical outcomes of castration-resis-
docetaxel resistance in metastatic castration-resistant prostate cancer. Eur. tant prostate cancer treatments administered as third or fourth line following
Urol. 2016; 70: 709–13. failure of docetaxel and other second-line treatment: results of an Italian
90 Tryggvadottir L, Vidarsdottir L, Thorgeirsson T et al. Prostate cancer pro- multicentre study. Eur. Urol. 2015; 68: 147–53.
gression and survival in BRCA2 mutation carriers. J. Natl Cancer Inst. 104 Dan TD, Eldredge-Hindy HB, Hoffman-Censits J et al. Hematologic toxicity
2007; 99: 929–35. of concurrent administration of radium-223 and next-generation antiandro-
91 Narod SA, Neuhausen S, Vichodez G et al. Rapid progression of prostate gen therapies. Am. J. Clin. Oncol. 2017; 40: 342–7.
cancer in men with a BRCA2 mutation. Br. J. Cancer 2008; 99: 371–4. 105 Michael JM, Yohann L, Karim F et al. Effects of radium-223 (Ra-223) with
92 Cheng HH, Pritchard CC, Boyd T, Nelson PS, Montgomery B. Biallelic docetaxel versus docetaxel alone on bone biomarkers in patients with bone-
inactivation of BRCA2 in platinum-sensitive metastatic castration-resistant metastatic castration-resistant prostate cancer (CRPC): a phase I/IIa clinical
prostate cancer. Eur. Urol. 2016; 69: 992–5. trial. J. Clin. Oncol. 2017; 35: 154.

© 2017 The Japanese Urological Association 231

You might also like