Download CRPC - Shifa International Hospitals

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Prostate-specific antigen wikipedia , lookup

Transcript
R EVI E W A R T IC L E
Targeting the androgen receptor
signalling axis in castration-resistant
prostate cancer (CRPC)
BJUI
BJU INTERNATIONAL
Che-Kai Tsao, Matthew D. Galsky, Alexander C. Small,
Tiffany Yee and William K. Oh
Division of Hematology and Medical Oncology, The Tisch Cancer Institute, Mount Sinai
School of Medicine, New York, NY, USA
Accepted for publication 31 May 2012
What’s known on the subject? and What does the study add?
Castration resistance has been appreciated for decades, and several mechanisms
theorising on this effect have been proposed. A rich pipeline of novel agents, including
abiraterone and MDV3100, have provided proof of principle that novel agents targeting
the AR signalling pathway with superior selectivity and activity than predecessors have
yielded significant clinical benefit for patients with metastatic castration-resistant
prostate cancer.
Our review provides an update in the development of several novel agents targeting the
AR signalling pathway now in clinical testing, as well as review novel therapies in
development with distinct mechanisms of action showing promising preclinical activity.
• Despite undergoing local therapy with curative intent, 20–30% of patients with prostate
cancer will ultimately development metastatic disease, leading to morbidity and mortality.
• Androgen-deprivation therapy (ADT) for men with metastatic prostate cancer results in
transient clinical benefit, but ultimately, cancers progress despite castrate levels of serum
testosterone, a clinical state classically referred to as ‘hormone refractory’ disease.
• In this review, we examine mechanisms of resistance to ADT that have redefined our
understanding of the more appropriately termed ‘castration resistant’ disease, and have
paved the way for a new generation of therapeutics targeting the androgen signalling axis
in advanced prostate cancer.
KEYWORDS
INTRODUCTION
prostate cancer, castration resistance, androgen deprivation, novel endocrine therapy,
hormone refractory, androgen receptor
Prostate cancer is the most common non-skin cancer in men in the Western world [1]. While
widespread PSA screening has significantly increased prostate cancer detection, about
one-third of men treated with definitive local therapy will have disease recurrence, while
another subset will present with advanced disease at the time of initial diagnosis [2].
Prostate cancer growth and progression is stimulated by androgens, acting through the
androgen receptor (AR). Circulating androgen levels are predominately regulated through the
hypothalamic-pituitary-adrenal-gonadal axis and androgen-deprivation therapy (ADT) results
in temporary disease regression in most patients. However, this approach eventually fails, as
tumours develop mechanisms to grow despite chemical or surgical castration and inevitably
leading to significant morbidity and mortality.
THE NORMAL PROSTATE AND THE AR
‘Prostate cancer growth and progression is
stimulated by androgens, acting through the
androgen receptor’
1580
The prostate is responsible for producing
most of the fluid in the seminal plasma
volume of men’s ejaculate. Binding of
©
2 0 1 2 B J U I N T E R N A T I O N A L | 11 0 , 1 5 8 0 – 1 5 8 8 | doi:10.1111/j.1464-410X.2012.11445.x
TARGETING THE AR SIGNALLING AXIS IN CRPC
FIG. 1.
AR signalling and mechanisms
of castration resistance.
Testes
MECHANISMS OF CASTRATION RESISTANCE
Steroidogenesis
Ligand Dependent Ligand Independent
Adrenal gland
Persistent
intratumoral
androgens
DHT
AR
AR Splice
Variants
Autocrine
Androgen
Production
AR
AR
AR
AR
AR
Gene
Amplification
Tumour cell
proliferation
PROSTATE TUMOUR CELL
AR Mutation
Co-activators
Co-repressors
AR
Proteins
CYTOPLASM
NUCLEUS
RNA
AR
Epigenetic
changes
androgens to the AR, expressed in epithelial
and stromal cells of the prostate [3], results
in differentiation, metabolism, proliferation,
and survival of these cells. The AR is
composed of ligand-dependent intracellular
transcription factors that are known to
influence the development and growth of
prostate cancer. Mouse models show that
androgens are required at every step of
prostate development [4], as a nonfunctional AR results in a testicular
feminisation-like syndrome with absence of
the prostate [5]. While androgens are
produced by both the Leydig cells of the
testes (testosterone: 90–95%) and adrenal
gland (5-dehydroepiandrosterone [DHEA]
and androstenedione: 5–10%), conversion to
dihydrotestosterone (DHT) by 5α-reductase
occurs in the prostate.
The AR has three main regions: the
N-terminal domain (NTD), which contains
the activation function; a central DNA
binding domain; and a COOH-terminal
domain (CTD), which contains the ligandbinding region. The AR is a nuclear hormone
receptor normally bound in a complex with
multiple chaperones (heat-shock proteins,
hsps). Upon androgen binding, AR changes
its conformation, leading to nuclear
©
2012 BJU INTERNATIONAL
translocation and subsequent dimerization
with androgen response elements in the
promoter and enhancer regions of target
genes [6]. Through cytoplasmic signalling
[7–9] and recruitment of co-activator
proteins [10], target gene transcription is
enhanced, which leads to cellular
proliferation.
PROSTATE CANCER: THE ROLE OF
ANDROGEN AND AR
clones exist before the initiation of ADT is
not known [11]. With ADT, selective pressure
selects for androgen-resistant clones
preferentially to proliferate. Additional
molecular changes may cause resistant
populations of cells to become dominant,
leading to state known as castrationresistant prostate cancer (CRPC). Several
mechanisms of castration resistance have
been proposed, classified broadly as either
ligand-dependent or ligand-independent
pathways (Fig. 1). The clinical management
of patients with CRPC is challenging, as
several of these mechanisms may be
involved simultaneously in any given
patient.
LIGAND-DEPENDENT MECHANISMS OF
CASTRATION RESISTANCE
Persistent androgens despite castration
Despite castration, alterations in the
multi-step processing of androgen synthesis
and transport can provide high enough
concentrations of androgens in the tumour
microenvironment to drive tumour growth
and progression. The clinical efficacy of
secondary hormonal agents targeting the
androgen–AR axis after progression despite
castration has provided a proof of principle
Since the work of Huggins and Hodges
demonstrated regression of metastatic
prostate cancer
with surgical
‘AR is a nuclear hormone receptor normally
castration in the
bound in a complex with multiple chaperones’
1940s, manoeuvres
to deplete
that residual androgens can stimulate
androgens have remained the standard
continued tumour proliferation. Both
first-line treatment for metastatic disease.
adrenal and intra-tumoral androgens have
However, this strategy ultimately fails as
been recognised to play an essential
tumours develop mechanisms to grow
role in the development of CRPC [12].
despite low levels
of circulating
androgens.
Whether resistant
prostate cancer
‘Both adrenal and intra-tumoral androgens
have been recognised to play an essential role
in the development of CRPC’
1581
TSAO ET AL.
Polymorphisms in androgen conversion and
transportation have also been associated
with outcomes in CRPC [13]. Furthermore, a
recent study showed that DHT synthesis
may bypass testosterone in driving
castration resistance [14]. Taken together,
these findings highlight the multiple steps in
the pathway of androgen synthesis and
transport that can be commandeered by
tumour cells to maintain adequate levels of
androgens in the microenvironment to
support growth.
The adrenal glands have long been
recognised as a potential source of
androgen production despite surgical or
chemical castration. Early attempts to
maximize adrenal androgen blockade via
adrenalectomy had limited efficacy [15].
Subsequently, the antifungal ketoconazole, a
cytochrome P450 and 17,20-lyase inhibitor
blocking the synthesis and degradation of
adrenal steroids, was used to treat men with
CRPC confirming the importance of
extra-testicular sources of androgen
production in subsets of patients [16]. The
clinical activity of ketoconazole also
provided the conceptual, and molecular,
framework for the development of more
selective adrenal androgen inhibitors.
More recently, the importance of
intratumoral androgen production has
emerged. Several studies showed that
despite castrate levels of serum testosterone,
residual levels of testosterone remain
sufficiently elevated in the prostate cancer
microenvironment, suggesting an ‘autocrine’
pathway of androgen production may play
an important role in developing castration
resistance [12]. Selective stress from
castration causes prostate cancer to
up-regulate enzymes responsible for de novo
steroidogenesis and adrenal steroid
conversion and fuel intra-prostatic androgen
[17–21].
AR gene amplification
‘epigenetics
1582
Compared with hormone-sensitive prostate
cancer cells, up to one-third of CRPC
tumours harbour
AR gene
may play an important role in the amplification
[22–24]. Despite
development of CRPC’
castration, AR
hypersensitivity to
low levels of androgens results in continued
tumour proliferation [23,25]. In addition,
heightened AR sensitivity to DHT is
associated with increased AR expression,
stability, and nuclear localisation in CRPC
cells [26]. Therapeutic use of AR blockade
may also lead to AR over-expression and
hypersensitivity [27].
LIGAND-INDEPENDENT MECHANISMS OF
CASTRATION RESISTANCE
Conventional hormonal therapy is ineffective
when tumours grow in a truly androgen
independent manner, thus posing a major
clinical challenge, particularly as more
potent and selective androgen biosynthesis
inhibitors and AR blockers emerge. Several
mechanisms have been hypothesised to play
important roles in the development of
castration resistance including genetic
variations, epigenetic changes, and alteration
of transcriptional and translational
regulation, leading to tumour proliferation
without ligand binding.
AR mutations
Although AR mutations are uncommon in
CRPC, it has been speculated that AR is
activated by several hormones, including
progesterone, oestrogen, adrenal androgens
and metabolic by-products of DHT [28–30].
In addition, mutant ARs may also
bind AR antagonists [31,32], as well as
corticosteroids [33]. The promiscuous nature
of the mutant AR may be responsible for
the anti-androgen withdrawal syndrome,
whereby patients with progressive disease
on an anti-androgen experience a transient
disease remission simply by discontinuing
the anti-androgen. In these cases, the drug
was presumably exerting agonistic, rather
than antagonistic functions [34].
Epigenetic modification
Recent work has shown that epigenetics
may play an important role in the
development of CRPC [35]. Preclinical
models have shown that post-translational
modifications, e.g. histone modification and
DNA methylation, can lead to castration
resistance [36]. DNA hypermethylation
down-regulates the AR suppressor binding
complex leading to increased AR expression
[37], while DNA hypomethylation during
androgen deprivation increases AR signalling
in CRPC [38]. Adaptor/scaffolding protein
©
2012 BJU INTERNATIONAL
TARGETING THE AR SIGNALLING AXIS IN CRPC
TABLE 1 Mechanisms of novel AR-targeted agents in clinical development
Mechanism of castration resistance
Ligand dependent
Residual androgens (adrenal + intratumoral)
AR binding (gene amplification)
Ligand independent
Constitutively active AR splice variants
Epigenetic modifications
Drug action
Novel drugs
CYP17 inhibition
17,20-lyase inhibitor
Analogue of 3β-androstanediol (d-cholesterol -x- > d-pregnenolone)
CYP17 inhibition + AR antagonist
AR antagonist + AR translocation inhibitor
Analogue of 3β-androstanediol (AR antagonist)
CYP17 inhibition + AR antagonist
Targets AR NTD
Abiraterone
TAK-700
Apoptone
TOK-001
MDV3100/ARN509
Apoptone
TOK-001
EPI-001
AR antagonist + AR translocation inhibitor
Anti-AR mRNA
HDACi
Hypomethylating agent
MDV3100
EZN4176
Panobinostat
Azacitidine
TABLE 2 Recent and on-going Phase III clinical trials of novel agents targeting AR signalling axis
Clinical Trial
COU-301
COU-302
Agent
Abiraterone
Abiraterone
No. of
patients
1000
1158
Criteria
Post-Docetaxel
Pre-Docetaxel
Primary
endpoint
OS
OS
AFFIRM
PREVAIL
NCT01193244
NCT00716794
MDV3100
MDV3100
TAK-700
TAK-700
1199
1680
1083
1454
Post-Docetaxel
Pre-Docetaxel
Post-Docetaxel
Pre-Docetaxel
OS
OS, PFS
OS, PFS
OS, PFS
Outcome/primary
completion date
Favourable OS
Favourable PFS
Study Un-blinded
Favourable OS
September, 2014
October, 2013
January, 2013
NOVEL ENDOCRINE THERAPIES
PFS, progression-free survival; OS, overall survival.
receptor for activated C kinase 1 (RACK1),
via binding to the tyrosine kinase Src,
modulates the tyrosine phosphorylation of
AR, facilitating AR translocation. This
process is ligand independent, resulting in
up-regulation of transcriptional activity in
hormonally treated prostate cancer cells
[39]. AR-mediated transcriptional activation
of several proliferation and apoptotic
pathways is critically dependent on histone
acetylation [40]. Novel therapeutic strategies
targeting these post-translational
modifications are now being investigated in
clinical trials, although early trials of these
agents have not yet shown significant
clinical activity [41,42].
AR splice variants
AR splice variants ARs have been
hypothesised to contribute to the
©
2012 BJU INTERNATIONAL
the setting of castration, co-activators such
as hsp27 [46,47], Her2Neu tyrosine kinase
[48], bcl-2 [49], and IGF-binding protein 5
[50] are up-regulated, and associated with
the development of castration resistance.
Likewise, co-repressors have been described
in AR regulation [51], with mechanisms such
as direct sequestration by DAX-1 [42], and
interruption of AR C- and N-terminal
interaction by Filamin-A [52,53].
development of CRPC [43]. Under normal
conditions, the androgen-AR complex will
translocate into the nucleus, and regulate
expression of androgen-responsive genes.
However, AR splice variant isoforms are
constitutively active, promoting tumour cell
growth independently of ligand [44]. Recent
work has shown that the AR signalling
inhibitor MDV3100 inhibits the growth of
prostate cancer cell lines harbouring some
AR splice variants [45].
AR co-activators and co-repressors
Upon binding with androgen, AR recruits
co-regulatory proteins, which include
chromatin-remodelling complexes and
transcription machinery that leads to
modulation of transcription. Several AR
co-activators have been identified as major
contributors to the development of CRPC. In
With a better understanding of the
mechanisms resulting in castration
resistance, novel therapeutic agents that
target residual androgen and the AR
signalling axis have shown significant
preclinical activity, and have rapidly entered
clinical testing (Table 1). Already, Zytiga
(abiraterone acetate) and Enzalutamide
(MDV3100) have both been shown to
improve survival in patients with CRPC
(Table 2).
Abiraterone acetate
Abiraterone acetate, an irreversible inhibitor
of cytochrome p450 complex CYP17, is an
oral agent that suppresses adrenal steroid
and intra-tumoral androgen synthesis.
Compared with its predecessor ketoconazole,
abiraterone is both more potent and
selective. In a phase I study, abiraterone
administered at multiple dose levels resulted
in marked reductions in serum testosterone
levels to <1 ng/dL, and resulted in regression
1583
TSAO ET AL.
of measurable disease [54]. Because
mineralocorticoids increased and resulted
in related adverse events (AEs), e.g.
hypertension, hypokalaemia, leg swelling,
adding epleronone was effective in
ameliorating these events. Responses
were also seen in patients previously
receiving ketoconazole in a second phase I
study [55].
‘evidence suggest that AR
common in CRPC and may
resistance to conventional
Two phase II trials using abiraterone
1000 mg and prednisone 10 mg daily
further demonstrated the favourable
tolerability and clinical activity in the
treatment of CRPC. In patients who were
chemotherapy naïve, 67% had a PSA level
decline of >50%, 37.5% of patients with
measurable disease
had partial
amplification is
response by RECIST
confer therapeutic criteria, and 66%
had stable disease
anti-androgens’
at 6 months [56].
However, secondary
mineralocorticoid excess resulted in
significant hypokalaemia (88%),
hypertension (40%), and fluid overload
(31%), necessitating eplerenone treatment. A
separate study evaluated patients with CRPC
that had previously received docetaxel [57].
Even in this heavily pre-treated population,
36% of the patients had >50% PSA level
declines, 18% of the patients had partial
radiographic response, and the median time
to PSA progression was 169 days.
Importantly, with the co-administration of
10 mg oral prednisone, clinical
manifestations of mineralocorticoid excess
were not observed.
Two large phase III randomised trials of
abiraterone were initiated to evaluate two
separate patient populations. COU-AA-301
randomised 1195 patients in a 2:1 ratio to
treatment with abiraterone and prednisone
in patients with CRPC who previously
received docetaxel. The combined treatment
arm showed a statistically significant overall
survival benefit (14.8 vs 10.9 months, hazard
ratio [HR] 0.65, P < 0.001) [58]. In addition,
other clinical endpoints, including time to
PSA progression (10.2 vs 6.6 months, HR
0.58, P < 0.001), radiographic progressionfree survival (5.6 vs 3.6 months, P < 0.001),
and PSA response (38% vs 10%, P < 0.001),
favoured the combined treatment arm.
Abiraterone was associated with increased
AEs, including fluid retention, hypokalaemia,
hypertension, transaminitis, and cardiac
1584
dysfunction in the combined treatment arm,
generally low-grade and manageable.
COU-AA-302 is a double-blinded, placebocontrolled, phase III study that randomised
1088 patients with chemotherapy naïve
CRPC to compare abiraterone 1000 mg and
prednisone 10 mg daily vs prednisone alone.
A recent press release of the interim analysis
by an independent monitoring committee
concluded that differences in radiographic
progression-free survival, overall survival,
and secondary endpoints favoured the
combined treatment arm [59].
MDV3100
Several lines of evidence suggest that AR
amplification is common in CRPC and
may confer therapeutic resistance to
conventional anti-androgens [60]. MDV3100
was developed based on preclinical activity
even in the presence of AR amplification
[61]. Compared with older generation
non-steroidal anti-androgens, MDV3100 has
greater affinity for AR, and in addition
inhibits AR nuclear translocation without
detectable agonist effects. In a phase I–II
study, escalating doses of MDV3100 were
evaluated in patients with progressive CRPC
[62]. A dose of 240 mg/day was selected for
further investigation, as higher doses
increased the risk of seizures. In the phase II
portion, administration of MDV3100
achieved PSA level declines of >50% in 56%
of patients and radiographic responses in
22% of patients. Circulating tumour cell
counts showed 49% (25/51) of patients
converting from an unfavourable count
before treatment (>5 cells/7.5 mL blood) to
a favourable count after treatment (<5
cells/7.5 mL blood) [63]. Interestingly, PET
imaging with 18F-fluoro-5α-DHT showed
decreased androgen to AR binding,
confirming the original hypothesised
mechanism of action. Two randomised,
placebo-controlled, phase III trials of
MDV3100 have been conducted, evaluating
its efficacy in patients with both pre(PREVAIL) and post-docetaxel CRPC
(AFFIRM). Recently reported interim analysis
of the AFFIRM trial, which randomised
1199 patients in a 1:1 ratio, showed
that estimated median survival was 18.4
months for men treated with MDV3100
compared with 13.6 months for men
treated with placebo (P < 0.001), with a
37% reduction in the risk of death with
MDV3100 [64].
©
2012 BJU INTERNATIONAL
TARGETING THE AR SIGNALLING AXIS IN CRPC
TAK-700
Epigenetic therapy
TAK-700 is a 17, 20-lyase inhibitor that
significantly reduces adrenal and testicular
androgen levels. After showing significant in
vitro activity against prostate cancer cell
lines, a phase I/II, dose-escalation study was
initiated. TAK-700 was administered at five
dose levels, with prednisone 5 mg twice
daily. With additional patients treated at a
dose of 400 mg twice daily, dose limiting
toxicity was seen. The most common AEs
were fatigue (17 patients, three grade ≥3
with 600 mg twice a day), nausea (11
patients, one grade 3), and vomiting (seven
patients, two grade ≥3). Pharmacokinetic
analysis showed dose proportional increases
in single and multiple dose maximum
plasma concentrations of the drug (Cmax)
and the area under the concentration–time
curve (AUC0–8h). Impressively, the median
testosterone and DHEA sulphate levels
decreased from 5.5 to 0.6 ng/dL and from
50.0 μg/dL to below quantifiable levels,
respectively. All patients treated with
≥300 mg doses had a PSA level decrease
and in patients who received at least three
cycles of TAK-700 ≥300 mg, 12 (80%) had
PSA level reductions of ≥50% and four
(27%) had reductions of ≥90%. Given its
efficacy and safety, two randomised,
placebo-controlled, phase III trials have been
initiated to evaluate TAK-700 with
prednisone in patients with metastatic CRPC
in the docetaxel-naïve and docetaxel-treated
populations.
Histone deacetylase inhibitors (HDACi), via
Galeterone (TOK-001) is the first compound
modulation of HDAC activity, have a broad
to have multi-functional inhibitory activity
spectrum of epigenetic activities that have
on the AR signalling axis. In addition to
yet to be fully defined. SAHA, also known as
inhibiting CYP17, TOK-001 is also a very
vorinostat, is a USA Food and Drug
potent inhibitor of adrenal androgens and
Administration
(FDA) approved
HDACi for the
‘Hypomethylating agents have also shown
treatment of
favourable preclinical activity in the treatment
cutaneous T cell
of CRPC’
lymphoma, and has
shown anti-tumour
activity in prostate
cancer cell lines [69]. In clinical trials as
ARs. A phase I, multicentre, dose-finding
monotherapy, vorinostat did not exhibit
study in patients with chemotherapy naïve
anti-tumour activity, and was associated
CRPC was published in the American Society
with significant AEs in men with docetaxelof Clinical Oncology (ASCO) meeting in 2012
treated CRPC [41]. A phase I trial of
[73]. Patients were enrolled in cohorts from
oral panobinostat, another HDACi, with
650 to 2600 mg of TOK-001 daily, with 36
or without docetaxel, showed that the
of 49 patients completing the 12-week
dose limiting toxicities were neutropenia
treatment course. The treatment was
and dyspnea, respectively [70]. As evident
generally well tolerated, with only one
in cell lines, adding panobinostat to
severe AE considered related to TOK-001
docetaxel showed clinical activity even in
(rhabdomyolysis with acute renal failure,
patients who previously progressed on
with high dose statin use). Overall 22% of
docetaxel, providing a suggestion that
treated patients had a >50% PSA level
HDACi may overcome chemotherapy
decline, and an additional 26% had 30–50%
resistance. A phase Ib study with i.v.
PSA level declines. Consistent with lyase
panobinostat with docetaxel is currently
inhibition, increased corticosteroids and
on-going.
suppressed androgens were seen with dose
escalation.
Hypomethylating agents have also shown
favourable preclinical activity in the
ARN-509
treatment of CRPC [71]. 5-Azacitadine, an
FDA-approved agent for the treatment of
Like its structural analogue of MDV3100,
myelodysplastic syndrome, is a potent false
ARN-509 inhibits both AR nuclear
substrate
competitive
inhibitor of
‘In addition to inhibiting CYP17, TOK-001 is
methyltransferase
also a very potent inhibitor of adrenal
via its incorporation
androgens and ARs’
into DNA and RNA
during cell
replication and transcription, leading to
translocation and AR binding to androgen
silencing of promoter genes. In mouse
response elements in DNA, but with greater
models, combined treatment with castration
efficacy in prostate cancer cells with
and azacitidine delayed time to castration
over-expressed AR [74]. In a pilot phase I
resistance [72]. In 36 patients with
study, 30 patients with metastatic CRPC
chemotherapy naïve CRPC, treatment
received ARN-509 daily in nine dose
with azacitidine exhibited acceptable
escalating cohorts [75]. The most common
treatment associated AEs (four grade
grades 1–2 treatment-related AEs were
3 fatigue, two grade 3 neutropenia,
fatigue (38%), nausea (29%), and pain
and no grade 4 AEs) but only modest
(24%), with one treatment-related grade 3
in vivo clinical activity [33]. A trial of
AE (abdominal pain) at 300 mg. At 12 weeks
combined therapy with docetaxel in patients of treatment, 42% of patients have had
with chemo-naïve CRPC is currently
≥50% PSA level declines. An optimal
underway.
biological dose of 240 mg daily was selected
Apoptone
Apoptone, also known as HE3235, is a novel
synthetic analogue of 3β-androstanediol
that has shown significant preclinical
activity against prostate and breast cancer
[65]. In cell lines, apoptone binds to ARs,
resulting in the down-regulation of Bcl-2
and increased expression of caspases. In
human prostate adenocarcinoma cell lines,
apoptone decreased AR expression, and in a
CRPC xenograft suppressed tumour growth
and tumoral androgen synthesis.
Additionally, independent of CYP17
inhibition, apoptone inhibits conversion of
d-cholesterol to d-pregnenolone. An
on-going phase I/II study is exploring the
pharmacokinetics, safety, and activity of
apoptone in patients with both chemo-naïve
and chemotherapy-treated metastatic CRPC
[66–68].
©
2012 BJU INTERNATIONAL
TOK-001
1585
TSAO ET AL.
for phase II investigation, with PSA response
at 12 weeks selected as the primary
endpoint [76].
REFERENCES
1
EZN4176
2
EZN4176, an anti-sense oligonucleotide
mRNA antagonist of AR, exhibited potent AR
and tumour inhibition in both androgen
dependent and independent xenograft
models. Upon administration, EZN-4176 is
hybridized and releases the complementary
sequences of AR mRNA, thus blocking
translation of the AR protein, leading to
inhibition of AR-induced tumour cell
growth, and promotes tumour cell apoptosis
in AR over-expressing tumour cells. This
therapy has now entered phase I clinical
testing.
EPI-001
While nearly all androgen ablative agents
target the CTD and its ligand binding region,
therapeutic effort against the NTD have
recently begun. EPI-001, a small molecule
that inhibits transactivation of the AR NTD,
has been developed as a promising agent
for the treatment of CRPC [77]. The unique
mechanism and target of this drug escape
several mechanisms of castration resistance
such as gain-of-function mutations in the
ligand-binding domain, or expression of
constitutively active splice variants. This
promising approach to targeting of the AR
has yet to enter clinical trials.
3
4
5
6
7
8
9
SUMMARY
For decades, treatment of CRPC has been
limited, resulting in dismal outcomes for
patients who failed initial hormonal
treatment. However, abiraterone and
MDV3100 have provided proof that despite
castration resistance, more potent and
selective agents targeting the androgen–AR
signalling axis can be effective. A rich
pipeline of novel therapeutic agents is
currently in various stages of preclinical and
clinical development. Continued efforts to
combat mechanisms of AR resistance will be
critical to improving prostate cancer
outcomes in the future.
10
11
12
CONFLICT OF INTEREST
13
Dr William K. Oh is a Consultant for Janssen,
Astellas, Medivation, Millenium, and receives
research funding from Millenium.
1586
Jemal A, Siegel R, Xu J, Ward E.
Cancer statistics, 2010. CA Cancer J Clin
2010; 60: 277–300
Ryan CJ, Elkin EP, Small EJ, Duchane
J, Carroll P. Reduced incidence of bony
metastasis at initial prostate cancer
diagnosis: data from CaPSURE. Urol
Oncol 2006; 24: 396–402
Litvinov IV, De Marzo AM, Isaacs JT. Is
the Achilles’ heel for prostate cancer
therapy a gain of function in androgen
receptor signaling? J Clin Endocrinol
Metab 2003; 88: 2972–82
Hayward SW, Cunha GR. The prostate:
development and physiology. Radiol Clin
North Am 2000; 38: 1–14
Ohno S. Major sex-determining genes.
Monogr Endocrinol 1978; 11: 1–140
Heinlein CA, Chang C. Androgen
receptor in prostate cancer. Endocr Rev
2004; 25: 276–308
Migliaccio A, Castoria G, Di Domenico
M et al. Steroid-induced androgen
receptor-oestradiol receptor beta-Src
complex triggers prostate cancer
cell proliferation. EMBO J 2000; 19:
5406–17
Kousteni S, Bellido T, Plotkin LI et al.
Nongenotropic, sex-nonspecific signaling
through the estrogen or androgen
receptors: dissociation from
transcriptional activity. Cell 2001; 104:
719–30
Castoria G, Lombardi M, Barone MV
et al. Androgen-stimulated DNA
synthesis and cytoskeletal changes in
fibroblasts by a nontranscriptional
receptor action. J Cell Biol 2003; 161:
547–56
Grossmann ME, Huang H, Tindall DJ.
Androgen receptor signaling in
androgen-refractory prostate cancer.
J Natl Cancer Inst 2001; 93: 1687–97
Isbarn H, Pinthus JH, Marks LS et al.
Testosterone and prostate cancer:
revisiting old paradigms. Eur Urol 2009;
56: 48–56
Nishiyama T, Hashimoto Y, Takahashi
K. The influence of androgen deprivation
therapy on dihydrotestosterone levels in
the prostatic tissue of patients with
prostate cancer. Clin Cancer Res 2004;
10: 7121–6
Yang M, Xie W, Mostaghel E et al.
SLCO2B1 and SLCO1B3 may determine
time to progression for patients
receiving androgen deprivation therapy
14
15
16
17
18
19
20
21
22
for prostate cancer. J Clin Oncol 2011;
29: 2565–73
Chang KH, Li R, Papari-Zareei M et al.
Dihydrotestosterone synthesis bypasses
testosterone to drive castration-resistant
prostate cancer. Proc Natl Acad Sci U S A
2011; 108: 13728–33
Bhanalaph T, Varkarakis MJ, Murphy
GP. Current status of bilateral
adrenalectomy or advanced prostatic
carcinoma. Ann Surg 1974; 179:
17–23
Witjes FJ, Debruyne FM, Fernandez del
Moral P, Geboers AD. Ketoconazole
high dose in management of hormonally
pretreated patients with progressive
metastatic prostate cancer. Dutch
South-Eastern Urological Cooperative
Group. Urology 1989; 33: 411–5
Fung KM, Samara EN, Wong C et al.
Increased expression of type 2 3alphahydroxysteroid dehydrogenase/type 5
17beta-hydroxysteroid dehydrogenase
(AKR1C3) and its relationship with
androgen receptor in prostate
carcinoma. Endocr Relat Cancer 2006;
13: 169–80
Titus MA, Schell MJ, Lih FB, Tomer
KB, Mohler JL. Testosterone and
dihydrotestosterone tissue levels in
recurrent prostate cancer. Clin Cancer
Res 2005; 11: 4653–7
Stanbrough M, Bubley GJ, Ross K
et al. Increased expression of genes
converting adrenal androgens to
testosterone in androgen-independent
prostate cancer. Cancer Res 2006; 66:
2815–25
Locke JA, Nelson CC, Adomat HH,
Hendy SC, Gleave ME, Guns ES.
Steroidogenesis inhibitors alter but do
not eliminate androgen synthesis
mechanisms during progression to
castration-resistance in LNCaP prostate
xenografts. J Steroid Biochem Mol Biol
2009; 115: 126–36
Belanger B, Belanger A, Labrie F,
Dupont A, Cusan L, Monfette G.
Comparison of residual C-19 steroids in
plasma and prostatic tissue of human,
rat and guinea pig after castration:
unique importance of extratesticular
androgens in men. J Steroid Biochem
1989; 32: 695–8
Visakorpi T, Hyytinen E, Koivisto P
et al. In vivo amplification of the
androgen receptor gene and progression
of human prostate cancer. Nat Genet
1995; 9: 401–6
©
2012 BJU INTERNATIONAL
TARGETING THE AR SIGNALLING AXIS IN CRPC
23 Koivisto P, Kononen J, Palmberg C
et al. Androgen receptor gene
amplification: a possible molecular
mechanism for androgen deprivation
therapy failure in prostate cancer.
Cancer Res 1997; 57: 314–9
24 Bubendorf L, Kononen J, Koivisto P
et al. Survey of gene amplifications
during prostate cancer progression by
high-throughout fluorescence in situ
hybridization on tissue microarrays.
Cancer Res 1999; 59: 803–6
25 Feldman BJ, Feldman D. The
development of androgen-independent
prostate cancer. Nat Rev Cancer 2001; 1:
34–45
26 Gregory CW, Johnson RT Jr, Mohler JL,
French FS, Wilson EM. Androgen
receptor stabilization in recurrent
prostate cancer is associated with
hypersensitivity to low androgen. Cancer
Res 2001; 61: 2892–8
27 Kawata H, Ishikura N, Watanabe M,
Nishimoto A, Tsunenari T, Aoki Y.
Prolonged treatment with bicalutamide
induces androgen receptor
overexpression and androgen
hypersensitivity. Prostate 2010; 70:
745–54
28 Culig Z, Klocker H, Eberle J et al. DNA
sequence of the androgen receptor in
prostatic tumor cell lines and tissue
specimens assessed by means of the
polymerase chain reaction. Prostate
1993; 22: 11–22
29 Koivisto P, Kolmer M, Visakorpi T,
Kallioniemi OP. Androgen receptor
gene and hormonal therapy failure of
prostate cancer. Am J Pathol 1998; 152:
1–9
30 Fenton MA, Shuster TD, Fertig AM
et al. Functional characterization of
mutant androgen receptors from
androgen-independent prostate cancer.
Clin Cancer Res 1997; 3: 1383–8
31 Culig Z, Hoffmann J, Erdel M et al.
Switch from antagonist to agonist of
the androgen receptor bicalutamide is
associated with prostate tumour
progression in a new model system. Br J
Cancer 1999; 81: 242–51
32 Scher HI, Kelly WK. Flutamide
withdrawal syndrome: its impact on
clinical trials in hormone-refractory
prostate cancer. J Clin Oncol 1993; 11:
1566–72
33 Zhao XY, Malloy PJ, Krishnan AV et al.
Glucocorticoids can promote androgenindependent growth of prostate cancer
©
2012 BJU INTERNATIONAL
34
35
36
37
38
39
40
41
42
43
44
cells through a mutated androgen
receptor. Nat Med 2000; 6: 703–6
Kelly WK. Endocrine withdrawal
syndrome and its relevance to the
management of hormone refractory
prostate cancer. Eur Urol 1998; 34
(Suppl. 3): 18–23
Gao L, Alumkal J. Epigenetic regulation
of androgen receptor signaling in
prostate cancer. Epigenetics 2010; 5:
100–4
Perry AS, Watson RW, Lawler M,
Hollywood D. The epigenome as a
therapeutic target in prostate cancer.
Nat Rev Urol 2010; 7: 668–80
Wang LG, Ossowski L, Ferrari AC.
Androgen receptor level controlled by a
suppressor complex lost in an androgenindependent prostate cancer cell line.
Oncogene 2004; 23: 5175–84
Karpf AR, Bai S, James SR, Mohler JL,
Wilson EM. Increased expression of
androgen receptor coregulator MAGE-11
in prostate cancer by DNA
hypomethylation and cyclic AMP. Mol
Cancer Res 2009; 7: 523–35
Kraus S, Gioeli D, Vomastek T, Gordon
V, Weber MJ. Receptor for activated C
kinase 1 (RACK1) and Src regulate the
tyrosine phosphorylation and function
of the androgen receptor. Cancer Res
2006; 66: 11047–54
Nakayama T, Watanabe M, Suzuki H
et al. Epigenetic regulation of androgen
receptor gene expression in human
prostate cancers. Lab Invest 2000; 80:
1789–96
Bradley D, Rathkopf D, Dunn R et al.
Vorinostat in advanced prostate cancer
patients progressing on prior
chemotherapy (National Cancer Institute
Trial 6862): trial results and interleukin-6
analysis: a study by the Department of
Defense Prostate Cancer Clinical Trial
Consortium and University of Chicago
Phase 2 Consortium. Cancer 2009; 115:
5541–9
Holter E, Kotaja N, Makela S et al.
Inhibition of androgen receptor (AR)
function by the reproductive orphan
nuclear receptor DAX-1. Mol Endocrinol
2002; 16: 515–28
Hu R, Dunn TA, Wei S et al. Ligandindependent androgen receptor variants
derived from splicing of cryptic exons
signify hormone-refractory prostate
cancer. Cancer Res 2009; 69: 16–22
Dehm SM, Schmidt LJ, Heemers HV,
Vessella RL, Tindall DJ. Splicing of a
45
46
47
48
49
50
51
52
53
novel androgen receptor exon generates
a constitutively active androgen receptor
that mediates prostate cancer therapy
resistance. Cancer Res 2008; 68:
5469–77
Watson PA, Chen YF, Balbas MD et al.
Constitutively active androgen receptor
splice variants expressed in castrationresistant prostate cancer require
full-length androgen receptor. Proc
Natl Acad Sci U S A 2010; 107:
16759–65
Rocchi P, So A, Kojima S et al. Heat
shock protein 27 increases after
androgen ablation and plays a
cytoprotective role in hormonerefractory prostate cancer. Cancer Res
2004; 64: 6595–602
Rocchi P, Beraldi E, Ettinger S et al.
Increased Hsp27 after androgen ablation
facilitates androgen-independent
progression in prostate cancer via signal
transducers and activators of
transcription 3-mediated suppression of
apoptosis. Cancer Res 2005; 65:
11083–93
Craft N, Shostak Y, Carey M, Sawyers
CL. A mechanism for hormoneindependent prostate cancer through
modulation of androgen receptor
signaling by the HER-2/neu tyrosine
kinase. Nat Med 1999; 5: 280–5
Raffo AJ, Perlman H, Chen MW, Day
ML, Streitman JS, Buttyan R.
Overexpression of bcl-2 protects
prostate cancer cells from apoptosis in
vitro and confers resistance to androgen
depletion in vivo. Cancer Res 1995; 55:
4438–45
Miyake H, Nelson C, Rennie PS, Gleave
ME. Overexpression of insulin-like
growth factor binding protein-5 helps
accelerate progression to androgenindependence in the human prostate
LNCaP tumor model through activation
of phosphatidylinositol 3 -kinase
pathway. Endocrinology 2000; 141:
2257–65
Wang L, Hsu CL, Chang C. Androgen
receptor corepressors: an overview.
Prostate 2005; 63: 117–30
Loy CJ, Sim KS, Yong EL. Filamin-A
fragment localizes to the nucleus to
regulate androgen receptor and
coactivator functions. Proc Natl Acad Sci
U S A 2003; 100: 4562–7
Gross M, Liu B, Tan J, French FS,
Carey M, Shuai K. Distinct effects of
PIAS proteins on androgen-mediated
1587
TSAO ET AL.
54
55
56
57
58
59
60
61
62
gene activation in prostate cancer cells.
Oncogene 2001; 20: 3880–7
Attard G, Reid AH, Yap TA et al.
Phase I clinical trial of a selective
inhibitor of CYP17, abiraterone acetate,
confirms that castration-resistant
prostate cancer commonly remains
hormone driven. J Clin Oncol 2008; 26:
4563–71
Ryan CJ, Smith MR, Fong L et al.
Phase I clinical trial of the CYP17
inhibitor abiraterone acetate
demonstrating clinical activity in
patients with castration-resistant
prostate cancer who received prior
ketoconazole therapy. J Clin Oncol 2010;
28: 1481–8
Attard G, Reid AH, A’Hern R et al.
Selective inhibition of CYP17 with
abiraterone acetate is highly active in
the treatment of castration-resistant
prostate cancer. J Clin Oncol 2009; 27:
3742–8
Danila DC, Morris MJ, de Bono JS
et al. Phase II multicenter study of
abiraterone acetate plus prednisone
therapy in patients with docetaxeltreated castration-resistant prostate
cancer. J Clin Oncol 2010; 28: 1496–501
de Bono JS, Logothetis CJ, Molina A
et al. Abiraterone and increased survival
in metastatic prostate cancer. N Engl J
Med 2011; 364: 1995–2005
Johnson & Johnson, Inc. Study
Unblinded: ZYTIGA® (abiraterone acetate)
Plus Prednisone for Asymptomatic or
Mildly Symptomatic ChemotherapyNaïve Patients with Metastatic
Castration-Resistant Prostate Cancer.
Available at: http://www.jnj.com/
connect/news/all/study-unblindedzytiga-abiraterone-acetate-plusprednisone-for-asymptomatic-or-mildlysymptomatic-chemotherapy-naivepatients-with-metastatic-castrationresistant-prostate-cancer. Accessed 23
March 2012
Chen CD, Welsbie DS, Tran C et al.
Molecular determinants of resistance to
antiandrogen therapy. Nat Med 2004;
10: 33–9
Tran C, Ouk S, Clegg NJ et al.
Development of a second-generation
antiandrogen for treatment of advanced
prostate cancer. Science 2009; 324:
787–90
Scher HI, Beer TM, Higano CS et al.
Antitumour activity of MDV3100 in
castration-resistant prostate cancer: a
1588
63
64
65
66
67
68
69
70
71
72
phase 1–2 study. Lancet 2010; 375:
1437–46
Higano CS, Beer TM, Taplin E et al.
Antitumor activity of MDV3100 in
pre- and post-docetaxel advanced
prostate cancer: long-term follow-up of
a phase I/II study. J Clin Oncol 2011; 29
(Suppl. 7): abstract 134
Scher HI, Fizazi K, Saad F et al. Effect
of MDV3100, an androgen receptor
signaling inhibitor (ARSI), on overall
survival in patients with prostate cancer
postdocetaxel: results from the phase III
AFFIRM study. J Clin Oncol 2012; 30
(Suppl. 5): abstract LBA1
Ahlem C, Kennedy M, Page T et al.
17 -alkynyl 3 , 17 -androstanediol
non-clinical and clinical pharmacology,
pharmacokinetics and metabolism.
Invest New Drugs 2012; 30: 59–78
Montgomery RB, Morris M, Ryan C
et al. Results of preclinical and clinical
phase I/II open-label dose-ranging trial
with HE3235, a synthetic adrenal
hormone, in castrate-resistant prostate
cancer. Mol Cancer Ther 2009; 8
(Meeting Abstract Supplement): A221
Montgomery RB, Morris MJ, Ryan CJ
et al. HE3235, a synthetic adrenal
hormone, in patients with castrationresistant prostate cancer (CRPC):
results of phase I/II clinical trial. J Clin
Oncol 2010; 28 (Suppl.): 15s, abstracy
4674
Harbor BioSciences Inc. Harbor
BioSciences Apoptone® for Prostate
Cancer Shows A Partial Overall
Response-New Positive Phase I/Iia Data.
Weber R ed. Chief Financial Officer, San
Diego, California, USA: Harbor
BioSciences, Inc., 2010
Welsbie DS, Xu J, Chen Y et al. Histone
deacetylases are required for androgen
receptor function in hormone-sensitive
and castrate-resistant prostate cancer.
Cancer Res 2009; 69: 958–66
Rathkopf D, Wong BY, Ross RW et al.
A phase I study of oral panobinostat
alone and in combination with docetaxel
in patients with castration-resistant
prostate cancer. Cancer Chemother
Pharmacol 2010; 66: 181–9
Festuccia C, Gravina GL, D’Alessandro
AM et al. Azacitidine improves
antitumor effects of docetaxel and
cisplatin in aggressive prostate cancer
models. Endocr Relat Cancer 2009; 16:
401–13
Zorn CS, Wojno KJ, McCabe MT,
73
74
75
76
77
Kuefer R, Gschwend JE, Day ML.
5-aza-2 -deoxycytidine delays androgenindependent disease and improves
survival in the transgenic
adenocarcinoma of the mouse prostate
mouse model of prostate cancer. Clin
Cancer Res 2007; 13: 2136–43
Montgomery RB, Eisenberger MA,
Rettig M et al. Phase I clinical trial of
galeterone (TOK-001), a multifunctional
antiandrogen and CYP17 inhibitor in
castration resistant prostate cancer
(CRPC). J Clin Oncol 2012; 30 (Suppl.):
abstract 4665
Sawyers CL. New insights into the
prostate cancer genome and therapeutic
implications. Proceedings of the Prostate
Cancer Foundation Annual Scientific
Retreat, 2010 September 15–16,
2010; Washington DC; 2010. Available
at: http://www.pcf.org/atf/cf/
%7B7C77D6A2-5859-4D60-AF47132FD0F85892%7D/PCFStateScience
2010.pdf. Accessed 23 March 2012
Rathkopf DE, Morris MJ, Danila DC
et al. A phase I study of the androgen
signaling inhibitor ARN-509 in patients
with metastatic castration-resistant
prostate cancer (mCRPC). J Clin Oncol
2012; 30 (Suppl.): abstract 4548
Rathkopf DE, Shore N, Antonarakis ES
et al. A phase II study of the androgen
signaling inhibitor ARN-509 in patients
with castration-resistant prostate cancer
(CRPC). J Clin Oncol 2012; 30 (Suppl.):
abstract TPS4697
Andersen RJ, Mawji NR, Wang J et al.
Regression of castrate-recurrent
prostate cancer by a small-molecule
inhibitor of the amino-terminus domain
of the androgen receptor. Cancer Cell
2010; 17: 535–46
Correspondence: William K. Oh, Mount Sinai
School of Medicine, The Tisch Cancer
Institute, 1 Gustave L Levy Place, New York,
NY 10029, USA.
e-mail: [email protected]
Abbreviations: ADT, androgen-deprivation
therapy; AE, adverse event; AR, androgen
receptor; ASCO, American Society of Clinical
Oncology; CRPC, castration-resistant
prostate cancer; CTD, COOH-terminal
domain; DHEA, dehydroepiandrosterone;
DHT, dihydrotestosterone; FDA, USA Food
and Drug Administration; HDAC(i), histone
deacetylase (inhibitors); hsp, heat-shock
protein; NTD, N-terminal domain.
©
2012 BJU INTERNATIONAL