Vous êtes sur la page 1sur 8

Cancer/Radiothrapie 19 (2015) 220227

Disponible en ligne sur

ScienceDirect
www.sciencedirect.com

Review article

The androgen receptor for the radiation oncologist


Rcepteur aux andrognes : ce que loncologue radiothrapeute doit savoir
L. Quero a , F. Rozet b , P. Beuzeboc c , C. Hennequin a,
a

Oncologie-radiothrapie, hpital Saint-Louis, 1, avenue Claude-Vellefaux, 75475 Paris, France


Institut mutualiste Montsouris, 42, boulevard Jourdan, 75014 Paris, France
c
Institut Curie, 26, rue dUlm, 75005 Paris, France
b

a r t i c l e

i n f o

Article history:
Received 29 December 2014
Received in revised form 1st February 2015
Accepted 4 February 2015
Keywords:
Prostate cancer
Androgen receptor
Radiotherapy

a b s t r a c t
Androgen deprivation therapy is widely used in combination with radiotherapy for the treatment of
prostate cancer. The knowledge of the biology of the androgen axis could help the radiation oncologist
to combine both modalities in an efcient way. Moreover, new drugs have recently been approved and
their role in combination with radiation needs pre-clinical and clinical studies. This review summarized
the main data on the biology of androgen receptor and the potential implications for the physician.
Mechanisms of interactions between androgen deprivation therapy and radiotherapy are also presented
and discussed.
2015 Socit franaise de radiothrapie oncologique (SFRO). Published by Elsevier Masson SAS. All
rights reserved.

r s u m
Mots cls :
Cancer de la prostate
Rcepteur aux andrognes
Radiothrapie

La suppression andrognique est de plus en plus utilise en association avec la radiothrapie dans le cancer de prostate. La connaissance de la biologie des andrognes peut aider loncologue radiothrapeute
dans le maniement des associations dhormonothrapie et de radiothrapie. En outre, de nouveaux
mdicaments ont t rcemment mis sur le march dans le cadre du cancer de prostate rsistant
la castration. Leur emploi potentiel avec la radiothrapie doit tre valu dans le cadre dtudes prcliniques et cliniques. Cette revue rsume les connaissances acquises sur le rcepteur aux andrognes
et leurs implications potentielles pour le clinicien. Les mcanismes dinteraction entre la suppression
andrognique et la radiothrapie sont galement rsums.
2015 Socit franaise de radiothrapie oncologique (SFRO). Publi par Elsevier Masson SAS. Tous
droits rservs.

1. Introduction
Combination of androgen deprivation therapy and radiotherapy
is now the standard of care for prostate cancers patients of the
high-risk group and some of the intermediate group. If the clinical
benet of this association is now clearly demonstrated, the biological interactions between the hormonal treatment and radiotherapy
are not fully understood. Moreover, new hormonal agents as abiraterone and enzalutamide have been approved for the treatment
of castration-resistant prostate cancers. Their use in earlier phases

Corresponding author.
E-mail address: christophe.hennequin@sls.aphp.fr (C. Hennequin).

of the disease, particularly with radiotherapy, is more and more


discussed.
The aim of this review is to summarize the present knowledge of
the physiology of the androgen receptor and to discuss the potential
interactions between old and new hormonal drugs with radiation.
2. Prostate cancer and androgens
2.1. Prostate cancer biology
Prostate is a gland that synthesizes components of the seminal
uid, including proteases as prostate-specic antigen (PSA). In the
normal adult prostate, the androgen receptor is expressed in all
luminal cells and in some epithelial basal cells as well in stromal
cells.

http://dx.doi.org/10.1016/j.canrad.2015.02.008
1278-3218/ 2015 Socit franaise de radiothrapie oncologique (SFRO). Published by Elsevier Masson SAS. All rights reserved.

L. Quero et al. / Cancer/Radiothrapie 19 (2015) 220227

Prostate cancer develops from epithelial cells, but it is not clear


if it arises from basal or luminal cells [1]. However, the role of
stromal cells is essential to promote growth of prostate cancer,
through the secretion of various growth factors. More than 50%
of prostate cancers harbour a gene fusion between the promoter
of transmembrane protease, serine 2 (TMPRSS2) and portions of
the coding regions of E-twenty-six (ETS) transcription factors [2],
mainly erythroblast transformation-specic gene (ERG). TMPRSS2
is an androgen responsive gene (see below). The ETS family members are transcription factors, regulating cell proliferation, cell
migration, cell cycle control and apoptosis [3]. Genomic proling
revealed that mutations in the androgen receptor signalling pathway is much more altered than the others pathways [4].
2.2. Androgen dependency of prostate cancer
Androgen stimulation is fundamental for prostate cancer
growth. Androgen deprivation therapy has demonstrated more
than 60 years ago a dramatic effect on cancer progression [5].
Androgen deprivation therapy is obtained by surgical castration
or by the use of luteinizing hormone-releasing hormone (LH-RH)
agonists or antagonists. Androgen deprivation therapy is supposed
to reduce the serum testosterone level under 0.5 ng/mL. It is still
debated whether a level lower than 0.2 ng/ml improves the clinical
response rate and its duration. Combination of LH-RH agonists and
anti-androgens is called complete androgen blockade: its usefulness was not demonstrated in the treatment of metastatic prostate
cancer. The androgen receptor could induce directly cancer growth,
for example via the TMPRSS2-ETS fusion factor. But it can also cross
talks with numerous growth factors (transforming growth factor
[TGF], vascular endothelial growth factor [VEGF], insulin-like
growth factor [IGF], epithelial growth factor [EGF] and broblast
growth factor [FGF]). For instance, TGF cooperates with androgen receptor signalling to promote stromal cell growth. Androgen
deprivation therapy remains the principal treatment of relapse
after radiotherapy [6].
2.3. Androgens synthesis
Testosterone is the principal circulating androgen and is synthesized by the testis for the major portion. Less than 3% of the
circulating testosterone is bioavailable, most of it being bound
to different proteins, sex hormone-binding protein (SHBG) and
albumin, essentially. The remaining androgens in the bloodstream
(510%) are dehydroepiandrosterone (DHEA), androstenediol and
androstenedione: they are produced by adrenals or by peripheral
conversion of testosterone. Androgens production is regulated by
the hypothalamic-pituitary-gonadal axis. LH-RH is secreted by the
hypothalamus in pulses, thus stimulating luteinizing hormone (LH)
secretion, which acts on Leydig cells in the testis to induce androgen
production. Testosterone acts on hypothalamus through a negative
feedback to prevent LHRH release.
Steroids synthesis begins with mobilization by the steroidogenic acute regulatory protein (StAR) and cleavage by CYP11A1
of cholesterol in pregnenolone (Fig. 1). Subsequent metabolism
to mineralocorticoids, glucocorticoids, androgens or oestrogens
depends of the enzymatic equipment of the tissue. Androgens are
produced from the conversion of pregnenolone-like steroids by the
CYP17, a cytochrome P450 enzyme. CYP17 catalyzes hydroxylase
and lyase reactions: the hydroxylase activity is similar for pregnenolone and progesterone but the lyase activity is much more
potent for 17-OH-pregenolone than 17-OH-progesterone. Hence,
the synthesis of androgens is mainly performed via DHEA. Polymorphisms of the CYP17 gene have been associated with a higher
risk of prostate cancer and a worst prognosis of castration-resistant
prostate cancers [7,8].

221

DHEA and androstenedione, synthesised in adrenal glands, are


transformed in testosterone in the Leydig cells of the testis. Testosterone is released in the bloodstream and enters into the target
cell via passive diffusion through the plasma membrane, but
active transport by organic anion-transport polypeptides have been
described [9]. Testosterone is converted into target tissues (prostate
and a limited number of other tissues) in 5-dihydro-testosterone
by 5-alpha reductase (SRD5A1 or 2). There are two types of 5-alpha
reductase, depending on the target tissue, the type 2 being more
specic for prostatic epithelial cells.
3. The androgen receptor
The androgen receptor is a nuclear transcription factor and is a
member of the steroid receptor superfamily. The androgen receptor gene is located on the X-chromosome at position Xq11-12 and
contains eight exons that encode a protein of 919 amino acids.
Two isoforms of androgen receptor have been described: the predominant isoform B (110 kDa) and the less dominant isoform A
(80 kDa).
Like other members of the nuclear receptor superfamily, the
androgen receptor is composed of four domains (Fig. 2) [10]:
the transcriptional activation domain (exon 1: N-terminal
domain): this domain is necessary and sufcient for transcription activity. It harbours a variable number of highly repetitive
DNA sequences, such as CAG, coding for polyglutamin. The number of CAG repeats is correlated to the transcriptional activity of
the androgen receptor, the shorter CAG repeat length resulting in
a higher transcriptional activity. Racial groups with a high prevalence of prostate cancer had also more frequently a short CAG
repeats, suggesting that early tumorigenesis is dependent on a
more active androgen receptor. The N-terminal domain contains
also coregulators interaction domains;
the DNA-binding domain (exon 2 and 3) with two zinc-nger
regions;
a exible hinge region separates the DNA-binding domain from
the androgen-binding domain: it contains the nuclear localization signal, allowing the androgen receptor to interact with the
cytoskeleton and to migrate to the nucleus, after binding of
importin- [11];
the ligand-binding domain at the C-terminal end (exons 4-8),
containing an androgen-binding pocket.
The activation function AF-1 and AF-2 domains are required
for optimal transactivation. AF-1 undergoes folding when contacted by transcription factor TFIIF after binding of the AR on DNA,
enabling coregulators recruitment. AF2 is a coactivator-binding
site, unmasked after ligand binding.
Ligand-free androgen receptor is sequestered in the cytoplasm
bound to heat-shock proteins (HSP 70, 90 and p23), which protect
it from degradation. After binding of the ligand, a conformational
change of the androgen receptor occurs, which allows dissociation
of heat-shock proteins.
Testosterone and dihydrotestosterone can bind to the androgen receptor, but the latter forms a more stable complex and
is 310 times more potent. Binding of the ligand initiates an
intramolecular interaction between the N-terminal domain and
ligand-binding domain, termed N/C interaction [12]. The androgen receptor becomes phosphorylated and translocates into the
nucleus (Fig. 3). The androgen receptor is phosphorylated at
several sites by different kinases, including mitogen-activated protein kinase (MAPK), protein kinase C (PKC) and protein kinase
B (AKT/PKB). They interact with the androgen receptor, usually
in a ligand-dependent manner. Phosphorylation stabilizes the

222

L. Quero et al. / Cancer/Radiothrapie 19 (2015) 220227

Fig. 1. Synthesis of steroids. In blue: classical pathway cholesterol is transformed in pregnenolone, then to dehydroepiandrosterone (DHEA) by cytochrome P450 enzyme
CYP17A1. DHEA is converted to androstenedione through the action of 3-hydroxysteroid dehydrogenase (3HSD) enzymes. 3HSD1 is present in peripheral tissues such as
prostate, skin or breast and is ten-fold more potent than 3HSD2 expressed in adrenal, testes and ovaries. Androstenedione is converted in testosterone by 3HSD3 (testis)
or AKCR1C3 (peripheral tissues). Then 5-dihydro-testosterone (5-DHT) is produced by 5-reductase 1 and 2. In green: Backdoor pathway17-OH-progesterone are acted
on by 5-reductase and aldo-keto reductase 1C (AKR1C) prior to the lyase activity of CYP17A to obtain androsterone, which is converted to androstanediol. An oxidative
step, which could be made by different enzymes (17BHSD 6 or 10, RODH4, RDH5, etc.) is required to convert androstanediol to 5-DHT. In pink: alternative pathway (5androstenedione pathway) androstenedione is converted rst by 5-reductase to 5-androstenedione and then by AKR1C3 (or 17HSD) to 5-DHT, thus circumventing
testosterone as a required intermediate.

ligand/receptor complex and is required for translocation to the


nucleus [13]. The androgen receptor dephosphorylation by the protein phosphatase 2A can lead to loss of its transcriptional activity
[14].
The androgen receptor forms homodimers (and also heterodimers with others proteins) and binds to specic DNA regions,
termed androgen-responsive elements, in association with a series
of transcriptional coregulators. These androgen-responsive elements are located in the promoter regions of target genes and
serve to activate or repress transcription of specic androgenregulated genes. About 146 androgen-regulated genes have been
described [15]. In the mature gland, androgens promote cell division and proliferation of epithelial cells, modulate programmed cell
death, regulate several aspects of prostate cellular metabolism and

control the production of specic proteins such as PSA. For example,


activation of androgen-responsive elements results in an enhanced
expression of cyclin-dependent kinases 2 and 4, down regulation
of the cell cycle inhibitor p16, thereby enhancing cell proliferation.
An up-regulation of p21, an inhibitor of apoptosis, is also observed.
4. Coregulators of the androgen receptor
By denition, these are proteins that increase or inhibit the
transcriptional activity of the androgen receptor [16]. Over 170
coregulators of androgen receptor functions have been identied,
either activators or repressors [17].
Some co-activators, as c-jun or steroid receptor co-activator-2
(src2), bind to the N-terminal domain of the androgen receptor and

Fig. 2. Structure of the androgen receptor and its gene. NTD: N-terminal domain (transcriptional domain); DBD: DNA-binding domain; LBD: ligand-binding domain; AF:
activation function.

L. Quero et al. / Cancer/Radiothrapie 19 (2015) 220227

223

Fig. 3. Androgens and androgen receptor signalling in prostate cancer. Dihydrotestosterone (DHT) binds to the androgen receptor (AR) and promotes its dissociation
from heat-shock protein (HSP). The androgen receptor translocates to the nucleus, dimerizes and binds to androgen-response elements (ARE), activating, with the help of
coactivators (COacT), androgen-regulated genes (ARG).

promote its homodimerization. SRC-2 is overexpressed or amplied


in 11% of prostate cancers [4]. SRC-2 expression is repressed by
androgens and thus, androgen deprivation therapy increases the
SRC-2 level.
Most coregulators are recruited to gene-promoter regions after
binding of androgen receptor dimers on androgen-responsive elements [12]. They function to facilitate or suppress transcription.
Various members of the steroids family can recognize the same
androgen-responsive element. So, the specic response of a tissue
is dependent on the coactivators or repressors. Some coactivators, including SRC-1 and cyclic adenosine monophosphate (cAMP)
response element-binding protein can induce the remodelling of
chromatin by acetylating histones, opening the nucleosomal structure of the gene and allowing the initiation of transcription by RNA
polymerase II.

receptor activation by a low concentration of androgens [19] or by


weak endogenous androgens. The androgen receptor could respond
to non-androgen steroidal hormones as progesterone and estradiol
[20]. Even anti-androgens such as utamide or bicalutamide could
become agonists of the androgen receptor [21]. However, these
mutations are detected in 10 to 20% of cancer specimens and are
responsible of the androgen escape in only a few patients [22,23].
Androgen receptor gene amplication is very common during
the castration-resistant state [24], leading eventually to its overexpression, making tumours cells extremely sensitive to low levels of
circulating androgens. Combined androgen blockade is potentially
a good way to overcome this androgen receptor amplication [25].

5. Mechanisms of castration resistance

Many genes are transcribed with alternative exon usage, and


this is the case for the androgen receptor gene. It has been demonstrated that alternative splicing of the androgen receptor mRNA
could culminate in a receptor that is capable of translocation and
DNA binding without ligand binding [26]. Expression of these variants is correlated with castration resistance [27]. AR-V7 is one of the
best-characterized splice variants, present in castration-resistant
prostate cancers patients [28]: it lacks the ligand-binding domain
and is constitutively active, independently of any androgens. Other
splice variants are required to dimerize with full-length receptors for nuclear translocation and transcriptional activity, and so
remains dependent of androgens [29].
The transcript ARv567es, in which exons 5, 6, and 7 are deleted,
appears to be less dependent of full-length receptors for transcriptional activity. On the other hand, AR-V7 and Arv567es facilitate
full-length receptors nuclear localization in the absence of androgen [30]. It has been recently shown that the presence of this
variant is correlated to abiraterone and enzalutamide resistance
[31]. However, there is at this moment no evidence that these
splice variants are present in localized diseases and in hormonenaive prostate cancers. For locally advanced diseases, when a
combination of androgen deprivation therapy and new

In advanced cancer treated with castration, a clinical and biochemical response is observed in the majority of patients, but
eventually, a resistance state will emerge after a variable delay. Castration resistance is dened as a progression of the disease, either
biological or clinical, with a testosterone level less than 0.5 ng/mL.
Although the exact mechanisms of castration resistance are not
well understood, available data support the physical presence and
activity of the androgen receptor. Castration-resistant prostate cancer is resistant to androgen deprivation therapy but remains often
androgen receptor-dependent [18]. This is evidenced by the usual
rise in serum PSA levels at the time of prostate cancer progression.
5.1. Mutation, amplication or overexpression of the androgen
receptor
In a relatively large proportion of patients with castrationresistant prostate cancers, the androgen receptor is expressed,
overexpressed, mutated or amplied. Mutations of the receptor
have been described, particularly in the ligand-binding domain.
Specicity of the ligand-binding domain could be modify, allowing

5.2. Alternative splicing of the androgen receptor messenger RNA

224

L. Quero et al. / Cancer/Radiothrapie 19 (2015) 220227

anti-androgens is considered, looking for this characteristic


could be recommended.
5.3. Alterations of coactivators and others intracellular pathways
Modications of coactivators has also been described: they
could increase the androgen receptor transcriptional activity in the
presence of low ligand concentration or by altering the ligand specicity of the receptor, allowing antiandrogens and estrogens to act as
agonists [32]. ARA70 is often overexpressed in castration-resistant
prostate cancers and facilitates the activation of the androgen
receptor by non-androgenic molecules such as 17-estradiol (E2)
[33]. Interaction of SRC-1 with the androgen receptor results in its
to the same magnitude as that obtained by dihydrotestosterone
[34]. SRC-1 and -2 expression is increased in more than 60% of
castration-resistant prostate cancers. In conclusion, it is clear that
overexpression of androgen receptor coactivators contribute to
prostate cancer progression.
Activation of the androgen receptor could be a consequence of
activating the PI3K/AKT/mTOR pathway via IGF-1, KGF or EGF [35].
The androgen receptor activation can result also by binding of long
non-coding RNAs [36].
5.4. Modications of the androgen synthesis pathways
It has been demonstrated that signicant amount of dihydrotestosterone have been detected in castration-resistant
tumours [37]. Numerous studies suggest that synthesis of androgens occur de novo in castration-resistant prostate cancers. Genes
encoding enzymes involved in androgen metabolism (and particularly CYP17A1) are up regulating in castration-resistant prostate
cancer cells [38]. Testosterone and dihydrotestosterone are usually
produced from cholesterol via the classical pathway in adrenal
glands and testis. However, in castration-resistant prostate cancer
cells, different pathways (backdoor and alternative pathways) are
activated to produce dihydrotestosterone (see Fig. 1) [37,3941].
Intracrine conversion of adrenal weak androgens such as DHEA, 11OH- androstenedione or deoxycorticosterone in steroids activating
the androgen receptor has been also identied [42]. In prostate,
DHEA is rst converted to androstenedione by 3-hydroxysteroid
dehydrogenase (3HSD1). Following castration, adrenal androgen
androstenedione serum and tissue levels are unchanged and a
signicant amount of androstenedione is observed in castrationresistant prostate cancer cells [43]. The alternative pathway seems
to be predominant in these cells [41]. Androstenedione is also
converted in 11-OH-androstenedione by CYP11B1,2 then reduced
in 11-OH-5-dihydrotestosterone by 5-reductase: this molecule
could activate the wild type androgen receptor [44]. All these
enzymes are potential targets for new drugs.
To summarize, four different molecular states of prostate cancer
can be dened [45]:
endocrine androgen-dependent and androgen receptordependent: prostate cancer growth is driven by serum androgens
produced by the testis;
intracrine androgen-dependent and androgen receptordependent: androgens are produced within prostate cancer
cells;
androgen (ligand)-independent and androgen receptordependent: this is the case for androgen receptor variants;
androgen-independent and androgen receptor-independent.
6. New hormonal drugs
In castration-resistant prostate cancers, growth of cancer cells
is often always dependent of the androgen receptor. X-ray

crystallography and computer modelling allow the understanding


of the biochemical interaction between drugs and the androgen
receptor at the atomic level. This helps to design new drugs to
overcome castration resistance.
Residual levels of androgens can be found in cancer cells due
in part to an intratumoral synthesis. Ketoconazole inhibits the 17hydroxylase activities of CYP17A1: at high dose, it has been shown
to obtain PSA response but no survival benet could be demonstrated [46]. Abiraterone acetate (Zytiga ) is an oral irreversible
inhibitor of CYP17A1, one of the major enzymes involved in androgen synthesis (Fig. 3) [47]. CYP17A1 has both 17a-hydroxylase
and 17,20-lyase activities. However, this enzyme is also required
for synthesis of glucocorticoids. Low dose dexamethasone is
required to prevent reexive increase of adrenocorticotropic
hormone (ACTH) and mineralosteroid effects, as hypertension
[48]. Abiraterone acetate decreased the production of androgens
(androstenedione, DHEA, testosterone 5-dihydrotestosterone) by
the adrenal glands, prostate and tumour cells [49].
Clinical trials showed that abiraterone acetate induced a dramatic response in many patients with a castration-resistant
prostate cancers [50]. In a retrospective study, abiraterone acetate
was shown to be superior to ketoconazole in terms of PSA
response and survival [51]. Randomized trials have demonstrated
an improvement in survival in castration-resistant prostate cancers
either after or before docetaxel [52,53].
TAK-700 (orteronel) is an another CYP17A1 inhibitor,
more potent to inhibit CYP17,20 lyase function than CYP17hydroxylase. It does not require an association with prednisone
to prevent mineralocorticoid effects. However, recently presented
phase III studies could not demonstrated an improvement in
survival in castration-resistant prostate cancers and this drug will
be probably not approved by the US Food and Drug Administration
(FDA) and the European Medicines Agency (EMA).
Non-steroidal androgen receptor antagonists, such as utamide
and bicalutamide, have relatively low afnity for the receptor and could, in some cases, become agonists. They do not
inhibit ligand-induced nuclear translocation but recruits nuclear
co-repressors to block androgen-regulated gene transcription
[54]. Enzalutamide (formerly MDV3100-Xtandi ) is eight times
more potent than bicalutamide to inhibit the androgen receptor
[55]; it inhibits not only the binding of testosterone and dihydrotestosterone, but also receptor translocation and binding of
coactivators [56]. Importantly, enzalutamide remains antagonist
of the androgen receptor in overexpressing cells and mutant cell
lines, which convert bicalutamide to an agonist [55]. Enzalutamide
increased overall survival for patients with castration-resistant
prostate cancers either after or before treatment with docetaxel
[57,58].
ARN-509 is an androgen receptor antagonist, differing from
enzalutamide by only one atom [59]. It also prevents androgen
receptor translocation and DNA-binding. Clinical trials are ongoing. Galeterone (TOK-001) inhibits CYP17 at low concentration, but
at higher doses, it could inhibit the androgen receptor and even
facilitates its degradation [60].
All these androgen receptor antagonists are targeting the
ligand-binding domain, which is lost in splice variants. Some new
inhibitors, as EPI-001, are directed against the N-terminal domain
and are supposed to inhibit protein protein interactions [61].
Although abiraterone and enzalutamide provide a survival benet, resistance almost always develops. Several new antiandrogens
agents are being evaluated either in preclinical or clinical studies
[62].
Finally, docetaxel, which inhibits the dissociation of microtubules, prevents nuclear translocation of the androgen receptor
and decreases its intracellular level. Part of its action could be mediated by hormonal inhibition [63,64].

L. Quero et al. / Cancer/Radiothrapie 19 (2015) 220227

7. Interactions between hormonal treatments and


radiation
Androgen deprivation therapy is today the standard hormonal
treatment of prostate cancer. In locally advanced stages, the combination of androgen deprivation therapy and radiotherapy has
demonstrated better survival rates than one modality used alone.
Long-term androgen deprivation therapy is used for high-risk
prostate cancers and short-term therapy for intermediate risk cancers. In contrast with the number of clinical trials involved, only a
few preclinical studies, either in vitro or in vivo, have evaluated the
mechanisms of interaction between hormonal manipulations and
radiations. This could be due to the lack of good preclinical models
for localized disease.
Preclinical models implicated prostate cancer cell lines: some as
LAPC4 or LNCaP are androgen-sensitive, unlike PC3 or DU145 cell
lines which do not expressed the androgen receptor. LNCaP cell
line presents an androgen receptor mutation (T877A LBD), which
allows binding of progesterone or cortisol [65]. LAPC4 cells have a
wild type androgen receptor.
Androgen deprivation therapy by itself induced a major reduction in epithelial cells mainly by apoptosis and decreased cell
proliferation [66]. The LNCaP cell line expressed the androgen
receptor and was regularly used for in vitro studies of combination of androgen deprivation therapy and radiation. Androgen
deprivation therapy was achieved by culture in charcoal-stripped
serum-containing medium. After irradiation, an increase in the
apoptosis rate was observed for LNCaP cultured in medium without
androgens compared to complete medium, but without any difference in clonogenic cell survival, suggesting a shift in the modality
of cell death [67].
Hermann et al. evaluated the effect of goserelin, an LH-RH agonist, on two cell lines, LNCaP et PC3; only LnCaP expressed the
androgen receptor but both cell lines expressed LH-RH receptors, as 86% prostate cancers do [68,69]. No radiosensitization was
observed in both cell lines despite inhibition of the EGF receptor
pathway and cell proliferation.
We have observed that combined treatment with bicalutamide,
an antiandrogen, with radiation concomitantly or sequentially, had
a protective effect over radiation alone in LNCaP cell line. There was
no signicant effect of bicalutamide on DU145 cell line, which does
not expressed the androgen receptor [70]. High levels of bicalutamide had a cytostatic effect on LnCap blocking the cell cycle in
G1 phase; senescence was the main cell death pathway.
In vivo, in different models of prostate cancer xenografts, it
seems that a neoadjuvant androgen deprivation therapy is required
to obtain and maximize the radiosensitizing effect. With the
Shionogi model, Zietman et al. showed that the TCD50 (radiation dose required to control 50% of the tumours) decreased when
androgen deprivation therapy was added to radiation, particularly
if it was given 12 days before irradiation [71]. This effect was correlated to tumour volume regression, suggesting that the decrease
of the number of clonogenic cells explained this supra-additive
effect. In another model, neoadjuvant androgen deprivation therapy (3 days) increased the apoptotic rate compared to radiation
alone or to androgen deprivation therapy given at the same time
than radiation [72]. However, this effect was not found in case of
multiple fractions, suggesting that apoptosis is not the main type
of radiation effect on prostate cancer cells [73]. In a model using
R3327-G dunning rat prostate tumour cells, neoadjuvant androgen
deprivation therapy resulted in prolonged suppression of tumour
growth, compared to irradiation alone or adjuvant androgen deprivation therapy [74].
The number of clonogenic cells, rate of apoptosis but also
hypoxia have been discussed as possible factors of radiosensitivity. Hypoxia is frequent in prostate cancer tissues and maybe also

225

in benign tissues [75,76]. The outcome is negatively inuenced by


hypoxia in patients treated by prostatectomy or radiotherapy [77].
More recently, in a study of 247 patients, in which hypoxia was
measured before radiotherapy, a pO2 10 mmHg was an independent of biochemical control and local control [78].
Inhibition of hypoxia-inducible factor (HIF) 1, the main mediator
of hypoxia, increases the radiosensitivity of PC3 cell line [79]. Looking at the relationship with the androgen axis, hypoxia increases
androgen receptor activation in LNCap cells [80]; HIF1 could be
a coactivator of the androgen receptor [81]. Androgen deprivation therapy decreases the hypoxia-induced expression of VEGF
in androgen-dependent LNCap cell line but not in the androgenindependent PC3 cell line [82]. In Shionogi prostate tumour model,
androgen deprivation therapy decreases the hypoxic fraction from
30 to 2% [83]. Antiandrogens reduce the transcriptional activity
of HIF1 [84]. In one study of 22 patients, hypoxia was measured
using Eppendorf microelectrode technique, androgen deprivation
therapy increased the median pO2 values from 6.4 to 15 mmHg:
part of the radiosensitizing effect of androgen deprivation therapy could be explained by this way [85]. On the other hand, in
a study of 20 patients where dynamic magnetic resonance imaging (MRI) evaluated vascularisation, androgen deprivation therapy
induced a profound reduction in tumour blood ow and an increase
of hypoxia [86].
More recently, it has been demonstrated that ionizing radiations
activated the androgen receptor. Moreover, the androgen receptor upregulated a number of DNA repair genes, particularly genes
of the non-homologous pathway, because these genes contain the
androgen receptor binding sites in their enhancer sequences [87].
In contrast, castration reduced the Ku67 level, a protein essential
for non-homologous end-joining repair of double-strand breaks
[88]. By this way, the androgen receptor promotes radioresistance and androgen deprivation increases DNA-damage and slow
double-strand break repair [89]. New anti-androgen drugs (ARN
509, enzalutamide) enhanced DNA damage and slow repair of DNA
double-strand breaks.
Looking at these results, mechanisms of interaction between
androgen deprivation therapy and radiation are not fully elucidated. New drugs such as abiraterone and enzalutamide give new
opportunities to increase the effect of radiation on prostate cancer.
Preclinical and clinical studies of combination of these drugs with
radiation are urgently needed.
8. Conclusion
The androgen receptor remains the main target of prostate cancer either in locally or advanced disease, in castration dependant
or resistant conditions. Androgen deprivation therapy is an important tool to improve the effect of radiotherapy. The knowledge of
androgen receptor effects is important for the radiation oncologists
to manage at its best the combination of both modalities. New hormonal drugs bring new opportunities to increase the potential of
radiotherapy.
Disclosure of interest
C.H. Occasional involvements: advisory services to Astellas,
Janssen and Ipsen.
L.Q., F.R., P.B. have not supplied their declaration of conict of
interest.
References
[1] Wang ZA, Mitrofanova A, Bergren SK, Abate-Shen C, Cardiff RD, Califano A, et al.
Lineage analysis of basal epithelial cells reveals their unexpected plasticity and

226

[2]

[3]
[4]
[5]

[6]
[7]

[8]

[9]

[10]
[11]

[12]
[13]

[14]

[15]

[16]

[17]
[18]
[19]
[20]

[21]

[22]

[23]

[24]

[25]

[26]

[27]

[28]

[29]

[30]

L. Quero et al. / Cancer/Radiothrapie 19 (2015) 220227


supports a cell-of-origin model for prostate cancer heterogeneity. Nat Cell Biol
2013;15:27483.
Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, Sun XW, et al.
Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate
cancer. Science 2005;310:6448.
Gutierrez-Hartmann A, Duval DL, Bradford AP. ETS transcription factors in
endocrine systems. Trends Endocrinol Metab 2007;18:1508.
Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, et al. Integrative genomic proling of human prostate cancer. Cancer Cell 2010;18:1122.
Huggins C, Stevens R, Hodges C. Studies on prostatic cancer. II. The effects
of castration on advanced carcinoma of the prostate cancer. Arch Surg
1941;43:20923.
Quero L, Hennequin C. Medical treatment for biochemical relapse after radiotherapy. Cancer Radiother 2014;18:5404.
Stanford JL, Noonan EA, Iwasaki L, Kolb S, Chadwick RB, Feng Z, et al. A polymorphism in the CYP17 gene and risk of prostate cancer. Cancer Epidemiol
Biomarkers Prev 2002;11:2437.
Hamada A, Sissung T, Price DK, Danesi R, Chau CH, Shari N, et al. Effect of
SLCO1B3 haplotype on testosterone transport and clinical outcome in caucasian patients with androgen-independent prostatic cancer. Clin Cancer Res
2008;14:33128.
Yang M, Xie W, Mostaghel E, Nakabayashi M, Werner L, Sun T, et al. SLCO2B1 and
SLCO1B3 may determine time to progression for patients receiving androgen
deprivation therapy for prostate cancer. J Clin Oncol 2011;29:256573.
Tan ME, Li J, Xu HE, Melcher K, Yong EL. Androgen receptor: structure, role in
prostate cancer and drug discovery. Acta Pharmacol Sin 2015;36:323.
Ni L, Llewellyn R, Kesler CT, Kelley JB, Spencer A, Snow CJ, et al. Androgen
induces a switch from cytoplasmic retention to nuclear import of the androgen
receptor. Mol Cell Biol 2013;33:476678.
van Royen ME, van Cappellen WA, de Vos C, Houtsmuller AB, Trapman J. Stepwise androgen receptor dimerization. J Cell Sci 2012;125:19709.
Gioeli D, Ficarro SB, Kwiek JJ, Aaronson D, Hancock M, Catling AD, et al.
Androgen receptor phosphorylation. Regulation and identication of the phosphorylation sites. J Biol Chem 2002;277:2930414.
Yang CS, Vitto MJ, Busby SA, Garcia BA, Kesler CT, Gioeli D, et al. Simian virus
40 small t antigen mediates conformation-dependent transfer of protein phosphatase 2A onto the androgen receptor. Mol Cell Biol 2005;25:1298308.
Nelson PS, Clegg N, Arnold H, Ferguson C, Bonham M, White J, et al. The program
of androgen-responsive genes in neoplastic prostate epithelium. Proc Natl Acad
Sci U S A 2002;99:118905.
Heemers HV, Tindall DJ. Androgen receptor (AR) coregulators: a diversity of
functions converging on and regulating the AR transcriptional complex. Endocr
Rev 2007;28:778808.
Heinlein CA, Chang C. Androgen receptor (AR) coregulators: an overview.
Endocr Rev 2002;23:175200.
Seruga B, Ocana A, Tannock IF. Drug resistance in metastatic castrationresistant prostate cancer. Nat Rev Clin Oncol 2011;8:1223.
Gottlieb B, Beitel LK, Wu JH, Triro M. The androgen receptor gene mutations
database (ARDB): 2004 update. Hum Mutat 2004;23:52733.
Taplin ME, Bubley GJ, Shuster TD, Frantz ME, Spooner AE, Ogata GK, et al.
Mutation of the androgen-receptor gene in metastatic androgen-independent
prostate cancer. N Engl J Med 1995;332:13938.
Taplin ME, Bubley GJ, Ko YJ, Small EJ, Upton M, Rajeshkumar B, et al. Selection
for androgen receptor mutations in prostate cancers treated with androgen
antagonist. Cancer Res 1999;59:25115.
Suzuki H, Ueda T, Ichikawa T, Ito H. Androgen receptor involvement
in the progression of prostate cancer. Endocr Relat Cancer 2003;10:
20916.
Taplin ME, Rajeshkumar B, Halabi S, Werner CP, Woda BA, Picus J, et al. Androgen receptor mutations in androgen-independent prostate cancer: Cancer and
Leukemia Group B Study 9663. J Clin Oncol 2003;21:26738.
Edwards J, Krishna NS, Grigor KM, Bartlett JM. Androgen receptor gene amplication and protein expression in hormone refractory prostate cancer. Br J
Cancer 2003;89:5526.
Palmberg C, Koivisto P, Hyytinen E, Isola J, Visakorpi T, Kallioniemi OP, et al.
Androgen receptor gene amplication in a recurrent prostate cancer after
monotherapy with the nonsteroidal potent antiandrogen Casodex (bicalutamide) with a subsequent favorable response to maximal androgen blockade.
Eur Urol 1997;31:2169.
Sun S, Sprenger CC, Vessella RL, Haugk K, Soriano K, Mostaghel EA, et al. Castration resistance in human prostate cancer is conferred by a frequently occurring
androgen receptor splice variant. J Clin Invest 2010;120:271530.
Hornberg E, Ylitalo EB, Crnalic S, Antti H, Stattin P, Widmark A, et al. Expression of androgen receptor splice variants in prostate cancer bone metastases
is associated with castration-resistance and short survival. PLoS One 2011;6:
e19059.
Guo Z, Yang X, Sun F, Jiang R, Linn DE, Chen H, et al. A novel androgen receptor
splice variant is up-regulated during prostate cancer progression and promotes
androgen depletion-resistant growth. Cancer Res 2009;69:230513.
Watson PA, Chen YF, Balbas MD, Wongvipat J, Socci ND, Viale A, et al. Constitutively active androgen receptor splice variants expressed in castration-resistant
prostate cancer require full-length androgen receptor. Proc Natl Acad Sci U S A
2010;107:1675965.
Cao B, Qi Y, Zhang G, Xu D, Zhan Y, Alvarez X, et al. Androgen receptor
splice variants activating the full-length receptor in mediating resistance to
androgen-directed therapy. Oncotarget 2014;5:164656.

[31] Antonarakis ES, Lu C, Wang H, Luber B, Nakazawa M, Roeser JC, et al. AR-V7 and
resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med
2014;371:102838.
[32] Edwards J, Bartlett JM. The androgen receptor and signal-transduction pathways in hormone-refractory prostate cancer. Part 2: androgen-receptor
cofactors and bypass pathways. BJU Int 2005;95:132735.
[33] Miyamoto H, Yeh S, Wilding G, Chang C. Promotion of agonist activity of
antiandrogens by the androgen receptor coactivator, ARA70, in human prostate
cancer DU145 cells. Proc Natl Acad Sci U S A 1998;95:737984.
[34] Ueda T, Bruchovsky N, Sadar MD. Activation of the androgen receptor Nterminal domain by interleukin-6 via MAPK and STAT3 signal transduction
pathways. J Biol Chem 2002;277:707685.
[35] Culig Z, Hobisch A, Cronauer MV, Radmayr C, Trapman J, Hittmair A, et al. Androgen receptor activation in prostatic tumor cell lines by insulin-like growth
factor-I, keratinocyte growth factor, and epidermal growth factor. Cancer Res
1994;54:54748.
[36] Yang L, Lin C, Jin C, Yang JC, Tanasa B, Li W, et al. lncRNA-dependent mechanisms of androgen-receptor-regulated gene activation programs. Nature
2013;500:598602.
[37] Montgomery RB, Mostaghel EA, Vessella R, Hess DL, Kalhorn TF, Higano CS, et al.
Maintenance of intratumoral androgens in metastatic prostate cancer: a mechanism for castration-resistant tumor growth. Cancer Res 2008;68:444754.
[38] Stanbrough M, Bubley GJ, Ross K, Golub TR, Rubin MA, Penning TM, et al.
Increased expression of genes converting adrenal androgens to testosterone
in androgen-independent prostate cancer. Cancer Res 2006;66:281525.
[39] Cai C, Chen S, Ng P, Bubley GJ, Nelson PS, Mostaghel EA, et al. Intratumoral
de novo steroid synthesis activates androgen receptor in castration-resistant
prostate cancer and is upregulated by treatment with CYP17A1 inhibitors. Cancer Res 2011;71:650313.
[40] Locke JA, Guns ES, Lehman ML, Ettinger S, Zoubeidi A, Lubik A, et al. Arachidonic acid activation of intratumoral steroid synthesis during prostate cancer
progression to castration resistance. Prostate 2010;70:23951.
[41] Chang KH, Li R, Papari-Zareei M, Watumull L, Zhao YD, Auchus RJ, et al. Dihydrotestosterone synthesis bypasses testosterone to drive castration-resistant
prostate cancer. Proc Natl Acad Sci U S A 2011;108:1372833.
[42] Uemura M, Honma S, Chung S, Takata R, Furihata M, Nishimura K,
et al. 5alphaDH-DOC (5alpha-dihydro-deoxycorticosterone) activates
androgen receptor in castration-resistant prostate cancer. Cancer Sci
2010;101:1897904.
[43] Mohler JL, Gregory CW, Ford 3rd OH, Kim D, Weaver CM, et al. The androgen
axis in recurrent prostate cancer. Clin Cancer Res 2004;10:4408.
[44] Mostaghel E. Beyond T and DHT novel steroid derivatives capable of wild type
androgen receptor activation. Int J Biol Sci 2014;10:60213.
[45] Nelson PS. Molecular states underlying androgen receptor activation: a framework for therapeutics targeting androgen signaling in prostate cancer. J Clin
Oncol 2012;30:6446.
[46] Keizman D, Huang P, Carducci MA, Eisenberger MA. Contemporary experience
with ketoconazole in patients with metastatic castration-resistant prostate
cancer: clinical factors associated with PSA response and disease progression.
Prostate 2012;72:4617.
[47] Boissier E, Loriot Y, Vignot S, Massard C. [Abiraterone acetate (AA): current
guidelines of prescription of abiraterone]. Bull Cancer 2014;101:38893.
[48] Attard G, Reid AH, Yap TA, Raynaud F, Dowsett M, Settatree S, et al. Phase I
clinical trial of a selective inhibitor of CYP17, abiraterone acetate, conrms
that castration-resistant prostate cancer commonly remains hormone driven.
J Clin Oncol 2008;26:456371.
[49] Harshman LC, Taplin ME. Abiraterone acetate: targeting persistent androgen dependence in castration-resistant prostate cancer. Adv Ther 2013;30:
72747.
[50] ODonnell A, Judson I, Dowsett M, Raynaud F, Dearnaley D, Mason M, et al.
Hormonal impact of the 17alpha-hydroxylase/C(17,20)-lyase inhibitor abiraterone acetate (CB7630) in patients with prostate cancer. Br J Cancer
2004;90:231725.
[51] Peer A, Gottfried M, Sinibaldi V, Carducci MA, Eisenberger MA, Sella A,
et al. Comparison of abiraterone acetate versus ketoconazole in patients with
metastatic castration resistant prostate cancer refractory to docetaxel. Prostate
2014;74:43340.
[52] de Bono JS, Logothetis CJ, Molina A, Fizazi K, North S, Chu L, et al. Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med
2011;364:19952005.
[53] Ryan CJ, Smith MR, de Bono JS, Molina A, Logothetis CJ, de Souza P, et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl
J Med 2013;368:13848.
[54] Ryan CJ, Tindall DJ. Androgen receptor rediscovered: the new biology and targeting the androgen receptor therapeutically. J Clin Oncol 2011;29:36518.
[55] Tran C, Ouk S, Clegg NJ, Chen Y, Watson PA, Arora V, et al. Development of
a second-generation antiandrogen for treatment of advanced prostate cancer.
Science 2009;324:78790.
[56] Beuzeboc P, Benderra MA, de La Motte Rouge T. [Management of enzalutamide,
a new hormonal therapy]. Bull Cancer 2014;101:10712.
[57] Scher HI, Fizazi K, Saad F, Taplin ME, Sternberg CN, Miller K, et al. Increased
survival with enzalutamide in prostate cancer after chemotherapy. N Engl J
Med 2012;367:118797.
[58] Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS, et al.
Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med
2014;371:42433.

L. Quero et al. / Cancer/Radiothrapie 19 (2015) 220227


[59] Clegg NJ, Wongvipat J, Joseph JD, Tran C, Ouk S, Dilhas A, et al. ARN-509:
a novel antiandrogen for prostate cancer treatment. Cancer Res 2012;72:
1494503.
[60] Niraula S, Chi K, Joshua AM. Beyond castration-dening future directions in the
hormonal treatment of prostate cancer. Horm Cancer 2012;3:313.
[61] Andersen RJ, Mawji NR, Wang J, Wang G, Haile S, Myung JK, et al. Regression of
castrate-recurrent prostate cancer by a small-molecule inhibitor of the aminoterminus domain of the androgen receptor. Cancer Cell 2010;17:53546.
[62] Mohler ML, Coss CC, Duke 3rd CB, Patil SA, Miller DD, Dalton JT. Androgen receptor antagonists: a patent review (2008-2011). Expert Opin Ther Pat
2012;22:54165.
[63] Darshan MS, Loftus MS, Thadani-Mulero M, Levy BP, Escuin D, Zhou XK,
et al. Taxane-induced blockade to nuclear accumulation of the androgen
receptor predicts clinical responses in metastatic prostate cancer. Cancer Res
2011;71:601929.
[64] Zhu ML, Horbinski CM, Garzotto M, Qian DZ, Beer TM, Kyprianou N. Tubulintargeting chemotherapy impairs androgen receptor activity in prostate cancer.
Cancer Res 2010;70:79928002.
[65] Matias PM, Carrondo MA, Coelho R, Thomaz M, Zhao XY, Wegg A, et al.
Structural basis for the glucocorticoid response in a mutant human androgen
receptor (AR(ccr)) derived from an androgen-independent prostate cancer. J
Med Chem 2002;45:143946.
[66] Westin P, Stattin P, Damber JE, Bergh A. Castration therapy rapidly induces
apoptosis in a minority and decreases cell proliferation in a majority of human
prostatic tumors. Am J Pathol 1995;146:136875.
[67] Pollack A, Salem N, Ashoori F, Hachem P, Sangha M, von Eschenbach A, et al.
Lack of prostate cancer radiosensitization by androgen deprivation. Int J Radiat
Oncol Biol Phys 2001;51:10027.
[68] Halmos G, Arencibia J, Schally A, Davis R, Bostwick D. High incidence of receptors for luteinizing hormone-releasing hormone (LHRH) and LHRH receptor
gene expression in human prostate cancers. J Urol 2000;163:6239.
[69] Hermann R, Schwarten D, Fister S, Grundker C, Rave-Frank M, Nitsche M, et al.
No supra-additive effects of goserelin and radiotherapy on clonogenic survival
of prostate carcinoma cells in vitro. Radiat Oncol 2007;2:31.
[70] Quero L, Giocanti N, Hennequin C, Favaudon V. Antagonistic interaction
between bicalutamide (Casodex) and radiation in androgen-positive prostate
cancer LNCaP cells. Prostate 2010;70:40111.
[71] Zietman A, Prince E, Nakfoor B, Park J. Androgen deprivation and radiation
therapy sequencing studies using the shionogi in vivo tumor system. Int J
Radiat Oncol Biol Phys 1997;38:106770.
[72] Joon DL, Hasegawa M, Sikes C, Khoo V, Terry N, Zagars G, et al. Supraadditive
apoptotic response of R3327-G rat prostate tumors to androgen ablation and
radiation. Int J Radiat Oncol Biol Phys 1997;38:10717.
[73] Pollack A, Ashoori F, Sikes C, Joon DL, von Eschenbach A, Zagars G, et al. The early
supra-additive apoptotic response of R3327-G prostate tumors to androgen
ablation and radiation is not sustained with multiple fractions. Int J Radiat
Oncol Biol Phys 2000;46:1538.
[74] Kaminski JM, Hanlon AL, Joon DL, Meistrich M, Hachem P, Pollack A. Effect
of sequencing of androgen deprivation and radiotherapy on prostate cancer
growth. Int J Radiat Oncol Biol Phys 2003;57:248.

227

[75] Stewart GD, Ross JA, McLaren DB, Parker CC, Habib FK, Riddick AC. The relevance of a hypoxic tumour microenvironment in prostate cancer. BJU Int
2009;105:813.
[76] Parker C, Milosevic M, Toi A, Sweet J, Panzarella T, Bristow R, et al. Polarographic
electrode study of tumor oxygenation in clinically localized prostate cancer. Int
J Radiat Oncol Biol Phys 2004;58:7507.
[77] Vergis R, Corbishley CM, Norman AR, Bartlett J, Jhavar S, Borre M, et al. Intrinsic markers of tumour hypoxia and angiogenesis in localised prostate cancer
and outcome of radical treatment: a retrospective analysis of two randomised
radiotherapy trials and one surgical cohort study. Lancet Oncol 2008;9:34251.
[78] Milosevic M, Warde P, Menard C, Chung P, Toi A, Ishkanian A, et al. Tumor
hypoxia predicts biochemical failure following radiotherapy for clinically localized prostate cancer. Clin Cancer Res 2012;18:210814.
[79] Palayoor ST, Mitchell JB, Cerna D, Degraff W, John-Aryankalayil M, Coleman CN.
PX-478, an inhibitor of hypoxia-inducible factor-1alpha, enhances radiosensitivity of prostate carcinoma cells. Int J Cancer 2008;123:24307.
[80] Park SY, Kim YJ, Gao AC, Mohler JL, Onate SA, Hidalgo AA, et al. Hypoxia increases
androgen receptor activity in prostate cancer cells. Cancer Res 2006;66:51219.
[81] Horii K, Suzuki Y, Kondo Y, Akimoto M, Nishimura T, Yamabe Y, et al.
Androgen-dependent gene expression of prostate-specic antigen is enhanced
synergistically by hypoxia in human prostate cancer cells. Mol Cancer Res
2007;5:38391.
[82] Stewart RJ, Panigrahy D, Flynn E, Folkman J. Vascular endothelial growth
factor expression and tumor angiogenesis are regulated by androgens in
hormone responsive human prostate carcinoma: evidence for androgen
dependent destabilization of vascular endothelial growth factor transcripts.
J Urol 2001;165:68893.
[83] Skov K, Adomat H, Bowden M, Dragowska W, Gleave M, Koch CJ, et al. Hypoxia
in the androgen-dependent Shionogi model for prostate cancer at three stages.
Radiat Res 2004;162:54753.
[84] Mabjeesh NJ, Willard MT, Frederickson CE, Zhong H, Simons JW. Androgens
stimulate hypoxia-inducible factor 1 activation via autocrine loop of tyrosine
kinase receptor/phosphatidylinositol 3-kinase/protein kinase B in prostate
cancer cells. Clin Cancer Res 2003;9:241625.
[85] Milosevic M, Chung P, Parker C, Bristow R, Toi A, Panzarella T, et al. Androgen withdrawal in patients reduces prostate cancer hypoxia: implications for
disease progression and radiation response. Cancer Res 2007;67:60225.
[86] Alonzi R, Padhani AR, Taylor NJ, Collins DJ, DArcy JA, Stirling JJ, et al. Antivascular effects of neoadjuvant androgen deprivation for prostate cancer: an in vivo
human study using susceptibility and relaxivity dynamic MRI. Int J Radiat Oncol
Biol Phys 2011;80:7217.
[87] Goodwin JF, Schiewer MJ, Dean JL, Schrecengost RS, de Leeuw R, Han S, et al. A
hormone-DNA repair circuit governs the response to genotoxic insult. Cancer
Discov 2013;3:125471.
[88] Al-Ubaidi FL, Schultz N, Loseva O, Egevad L, Granfors T, Helleday T. Castration
therapy results in decreased Ku70 levels in prostate cancer. Clin Cancer Res
2013;19:154756.
[89] Polkinghorn WR, Parker JS, Lee MX, Kass EM, Spratt DE, Iaquinta PJ, et al. Androgen receptor signaling regulates DNA repair in prostate cancers. Cancer Discov
2013;3:124553.

Vous aimerez peut-être aussi