Progress of Molecular Targeted Therapies For Prostatecancers
Progress of Molecular Targeted Therapies For Prostatecancers
Progress of Molecular Targeted Therapies For Prostatecancers
Review
a r t i c l e i n f o a b s t r a c t
Article history: Prostate cancer remains the most commonly diagnosed malignancy and the second leading cause of cancer-
Received 5 October 2011 related deaths in men in the United States. The current standard of care consists of prostatectomy and radiation
Received in revised form 18 November 2011 therapy, which may often be supplemented with hormonal therapies. Recurrence is common, and many develop
Accepted 19 November 2011
metastatic prostate cancer for which chemotherapy is only moderately effective. It is clear that novel therapies
Available online 29 November 2011
are needed for the treatment of the malignant forms of prostate cancer that recur after initial therapies, such
Keywords:
as hormone refractory (HRPC) or castration resistant prostate cancer (CRPC). With advances in understanding
Prostate cancer of the molecular mechanisms of cancer, we have witnessed unprecedented progress in developing new forms
EGFR of targeted therapy. Several targeted therapeutic agents have been developed and clinically used for the treat-
IGF-1R ment of solid tumors such as breast cancer, non-small cell lung cancer, and renal cancer. Some of these reagents
VEGFR modulate growth factors and/or their receptors, which are abundant in cancer cells. Other reagents target the
Androgen downstream signal transduction, survival pathways, and angiogenesis pathways that are abnormally activated
Hormone resistant in transformed cells or metastatic tumors. We will review current developments in this field, focusing specifically
on treatments that can be applied to prostate cancers. Finally we will describe aspects of the future direction of
the field with respect to discovering biomarkers to aid in identifying responsive prostate cancer patients.
© 2011 Elsevier B.V. All rights reserved.
Contents
1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 141
2. Inhibitors of androgens and androgen receptor signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 141
3. Growth factors and growth factor receptors inhibitors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142
3.1. ErbB inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142
3.1.1. Monoclonal antibodies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 143
3.1.2. Small tyrosine kinase inhibitors (TKI) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 143
3.2. Inhibitors for insulin-like growth factor (IGF) receptors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 144
3.3. Platelet-derived growth factor inhibitors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 144
4. Anti-angiogenesis targeting therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 145
5. Other molecular targets for prostate cancer therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 146
5.1. Prostate-specific membrane antigen (PSMA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 146
5.2. Heat shock protein (Hsp) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 146
5.3. mTOR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 146
5.4. Histone deacetylase (HDAC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 147
6. New targets and biomarkers for prostate cancer therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 147
6.1. The F77 antigen . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 147
6.2. Serine peptidase inhibitor Kazal type 1 (SPINK1) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 147
6.3. Survivin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 147
7. Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 148
Acknowledgment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 148
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 148
⁎ Corresponding author at: 252 John Morgan Building, 3620 Hamilton Walk, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania,
Philadelphia, PA 19104-6082, USA. Tel.: +1 215 573 9256; fax: +1 215 898 2401.
E-mail address: [email protected] (H. Zhang).
0304-419X/$ – see front matter © 2011 Elsevier B.V. All rights reserved.
doi:10.1016/j.bbcan.2011.11.003
W. Fu et al. / Biochimica et Biophysica Acta 1825 (2012) 140–152 141
Fig. 1. Prostate cancer's progression and treatment. Prostate cancer patients with high risk are recommended to have surgery, radiation, and/or hormone therapies. Although these
treatments are efficient in general, some patients have recurrent diseases, at which stage current treatment options, mostly chemotherapy, are limited in terms of clinical outcome.
It is hoped that development in targeted therapies can provide more options for the recurrent and metastatic prostate cancers.
142 W. Fu et al. / Biochimica et Biophysica Acta 1825 (2012) 140–152
Fig. 2. Molecular targets and designed intervention in targeted therapies for prostate cancer. In addition to androgen/androgen receptor (AR) pathways, many growth receptors
(EGFR/HER2, IGFR, PDGFR, and VEGFR), and their downstream molecules, have been explored as the targets to block growth signaling of tumor cells or the angiogenesis process
that provides nutrients to cancer cells. Antibodies to some prostate cancer specific antigens (e.g. PSMA and the F77 antigen) have also been developed.
and the subsequent formation of dehydroepiandrosterone (DHEA) Biotechnology) in combination with prednisone for the treatment
and androstenedione [14]. As DHEA and androstenedione are andro- of metastatic CRPC in men who have received prior docetaxel
gens and precursors of testosterone, inhibition of CYP17 activity by chemotherapy.
abiraterone decreases circulating levels of testosterone. After i.p. in-
jection of 0.5 mmol/kg abiraterone acetate (the pro-drug), the plasma
3. Growth factors and growth factor receptors inhibitors
concentration of abiraterone reached more than 1 μM and remained at
about 0.4 μM after 24 h [15]. Potter et al. reported that the inhibitory
Multiple growth factors and growth factor receptors have been
activity (e.g. IC50) of abiraterone for hydroxylase and lyase activity
identified as critical regulatory proteins in signaling networks that
were both at 3–4 nM [16]. The IC50 is within the range of in vivo abir-
are common to many cancer cells. Novel agents currently in clinical
aterone pharmacological concentration and therefore sufficient to ob-
treatment are designed to targeted specific protein families such as
tain clinical activity.
the epidermal growth factor receptor (EGFR) family and the
Activity of abiraterone acetate as a single agent was apparent even
platelet-derived growth factor receptor (PDGFR) family.
in Phase I trials, resulting in significant decreases of serum PSA levels
(50% or more) in 55% CRPC patients with or without prior ketocona-
zole therapy [17], and in 57% of chemotherapy free but hormone ther- 3.1. ErbB inhibitors
apy resistant prostate cancer patients [18]. Ketoconazole was initially
developed as an antimycotic agent but later found to be a nonspecific The human EGFR family (HER/ErbB) receptors have been recog-
inhibitor of steroidogenic enzymes. This study has suggested that nized as a very important family of receptor tyrosine kinases, which
abiraterone is superior to ketoconazole in clinically activity. In Phase are frequently reported to have significant impacts on the cellular sig-
II trials, when combined with low-dose glucocorticoids such as pred- naling networks within many different solid tumors, including breast
nisone, abiraterone acetate caused significant PSA drop (50% or more) cancer, colon cancer, lung cancer and prostate cancer [23]. This family
in 36% patients with progressive metastatic CRPC who failed comprises four closely related receptors: EGFR (HER-1/ErbB1), HER2
docetaxel-based chemotherapy [19], and in 67% of chemotherapy (Neu/ErbB2), HER3 (ErbB3), and HER4 (ErbB4). These transmem-
free CRPC patients [20]. In Phase III studies that involved a total of brane glycoproteins contain an extracellular ligand binding domain
1195 patients, abiraterone acetate plus prednisone (797 patients), and an intracellular receptor tyrosine kinase (RTK) domain. It has
compared to placebo plus prednisone (398 patients), prolonged over- been reported that EGFR is overexpressed in 18–37% prostate cancers
all survival among patients with metastatic CRPC who had disease [24–26]. Recently, Neto et al. also reported a significant direct correla-
progression after docetaxel-based chemotherapy [21]. The median tion of HER2/neu over-expression with the risk of death and recur-
overall survival was 14.8 months in the abiraterone acetate plus pred- rence in prostate cancer [27]. HER2 is also associated with the
nisone group vs 10.9 months in the control groups [22]. As a result of activation of androgen receptor and androgen-induced PSA expres-
the successful phase III trial, the US Food and Drug Administration sion [28]. These studies indicate that targeted agents for ErbB recep-
(FDA) has recently approved abiraterone acetate (Zytiga, Cougar tors, including monoclonal antibodies and small molecule tyrosine
W. Fu et al. / Biochimica et Biophysica Acta 1825 (2012) 140–152 143
Fig. 3. Structure of small molecule agents in targeted therapies for prostate cancer.
kinase inhibitors, can potentially provide treatment options for pros- cells. Trastuzumab is the first FDA approved therapeutic drug targeting
tate cancer. ErbB receptors to treat metastatic breast and stomach cancer. As HER2
overexpression has been observed in prostate cancer patients, ranging
3.1.1. Monoclonal antibodies from 25% of untreated primary tumors to 78% of castrate metastatic tu-
Cetuximab (C225/Erbitux), a monoclonal antibody initially ap- mors, it is believed Trastuzumab could be effective to treat prostate can-
proved by the Food and Drug Administration (FDA) for colorectal can- cer [35]. An early study indicated that Trastuzumab might have activity
cer in 2004, directly binds to the extracellular domain of EGFR and on androgen-dependent but not androgen-independent tumor, and
blocks ligand (EGF) binding. In preclinical research, Karashima et al. combination of Trastuzumab with Paclitaxel leads to inhibition of both
reported that Cetuximab had significant anti-tumor effect in a murine type of tumors in xenografted animal models [36].
prostate cancer model, interfering with cell proliferation/angiogenesis Docetaxel may induce expression of HER2 in a human prostate
and enhancing apoptosis [29]. Cetuximab is effective for some but not cancer model, especially in hormone independent tumor cells [37],
all prostate cancer model systems. For example, Cetuximab caused a suggesting a mechanism for acquired resistance to chemotherapies.
significant growth inhibition by inducing cell apoptosis in Du145 Thus, HER2 targeted therapies may provide an approach to overcome
cells, but not in PC3 cells [30]. The activity of Cetuximab is associated the chemo-resistance. However, it has to be investigated if the doce-
with its effective inhibition of phosphorylation of EGFR at sites Tyr- taxel effect is only limited to prostate cancer. Nevertheless, it seems
845 and Tyr-1173 in Du145 but not in PC3 cells. This result might plausible that a docetaxel/trastuzumab combination may become an
be related to higher EGFR expression level in Du145 than PC3. A effective therapeutic approach for hormone refractory prostate can-
fully humanized EGFR mAb, Panitumumab (Vectibix), demonstrates cer. In fact, a phase I study has already demonstrated the safety of ad-
higher affinity to EGFR than Cetuximab does and shows significant ministration of docetaxel, estramustine, and trastuzumab in patients
growth inhibition of PC3 cells in vitro [31]. In a phase I clinical with metastatic androgen-independent prostate cancer [38].
study, 3 out of 21 prostate cancer patients showed stable disease
after Panitumumab treatment [32]. 3.1.2. Small tyrosine kinase inhibitors (TKI)
Wagener et al. showed that Cetuximab increased the treatment ef- A class of tyrosine kinase inhibitors has been developed to damp-
fects of radiation for prostate cancer in vitro and in vivo [33]. In a en the activity of ErbB receptors [39]. Clinical development of small
phase Ib/IIa trial, Cetuximab in combination with doxorubicin were molecule inhibitors of ErbB receptors, including gifitinib (IRESSA/
administered weekly for 6 times in patients with metastatic CRPC. ZD1839), erlotinib, and lapatinib (Fig. 3), has also been explored in
The median survival of patients under the combination therapy was the treatment of patients with prostate cancer. Bonaccorsi et al.
longer compared to historical control groups, although minimal PSA reported that gefitinib, which affects EGF-stimulated activation of
declines were observed [34]. PI3K/Akt pathway, suppressed invasion and proliferation of
Trastuzumab (Herceptin) is a recombinant humanized monoclonal androgen-independent prostate cancer cell lines PC3 and DU145
antibody that targets HER2, another member of the ErbB family thought [40]. Vicentini and coworkers showed that gefitinib caused cell cycle
to play a role in regulating cell proliferation and differentiation of cancer arrest and initiation of apoptosis in both androgen-sensitive cells
144 W. Fu et al. / Biochimica et Biophysica Acta 1825 (2012) 140–152
(ND1, LNCaP and ALVA-31) and androgen-independent cells [41]. Ef- that regulate and propagate IGF activity in various tissues [57]. Epide-
fects of gefitinib on LNCaP, PC3 and DU145 cell lines was investigated miological studies indicate that circulating IGF-I levels are positively
by Sgambato [42], who found that the anti-proliferative effect of gefi- correlated with increased risk of prostate cancer [58]. Cardillo et al.
tinib was mainly cytostatic and associated with a block in the G0/G1 analyzed 43 paraffin-embeded prostate cancer samples and found ki-
phase of the cell cycle. In DU145 cells, this block was associated netic changing of IGF system as prostate tissue progressed from a nor-
with an increase in expression of the CDK inhibitor p27kip1 that mal to malignant state, suggesting that differential expression of IGF
was not evident in the LNCaP and PC3 cells. may be associated with the malignancy of tumor phenotype [59].
Little evidence exists suggesting that gefitinib as a single-agent Given the substantial evidence that the IGF pathway plays a role in
has activity in CRPC. Although phase I studies reported improved prostate cancer, interference with this pathway appears to be a po-
pain control, an open-label, single-arm phase II trial of gefitinib in pa- tential approach for targeted therapies. Cixutumumab (IMC-A12),
tients with advanced CRPC showed that single-agent gefitinib had no which is currently in Phase II for prostate cancer [60], is a monoclonal
clinical efficacy assessed by PSA response rate [43]. This outcome is in antibody directed against IGF-1R. The antibody selectively binds to
agreement with results from two other phase II trials with HRPC pa- the membrane-bound IGF-I receptor, thereby down-regulating the
tients, one involving 40 patients who had not received prior chemo- PI3K/AKT survival pathway. In human tumor xenograft models,
therapy and the other involving 58 patients in the US [44, 45]. Both blocking of IGF-IR by IMC-A12 resulted in rapid and profound growth
trials showed no response to gefitinib treatments by monitoring PSA inhibition of cancers of the breast, lung, colon and pancreas [61]. Wu
levels. Another EGFR TKI, Erlotinib, shows moderate activity in et al. found that IMC-A12 was effective in both androgen-dependent
CRPC, leading to reduction of PSA levels in some patients [46] or (AD) and androgen-independent (AI) variant human prostate cancer
delay of PSA rise in others [47]. Lapatinib, an inhibitor for both EGFR xenografts [62]. Interestingly, IMC-A12 treatment induces both G1 ar-
and HER2, also showed single agent activity in CRPC [48]. Unexpect- rest and apoptosis in the AD tumors, whereas G2-M arrest was the
edly, lapatinib showed no activity in early stage prostate cancer pa- predominant cell cycle effect seen in AI tumors. These studies indicate
tients [49, 50]. that IGF-IR may play distinct roles in the growth and maintenance of
The conflicting results of TKIs in prostate cancer might be traced to AD and AI prostate cancers [56].
the lack of sensitive forms of kinases in the patient population. In As IGF-IR signaling is known to mediate resistance to cytotoxic
non-small cell lung cancer (NSCLC), the response rate to EGFR tyro- chemotherapy and radiation, targeted disruption of the IGF-IR axis
sine kinase inhibitors was significantly increased when treatment combined with conventional cancer treatments may be more effec-
was administered to patients with tumors containing somatic EGFR tive in the inhibition of tumor growth. This idea is supported by the
mutations [51]. It is worth noting that in all the prostate cancer trials finding that IMC-A12 enhances the therapeutic effect of docetaxel
as discussed above, patients were not examined for inhibitor sensitive on advanced prostate cancer in animal models [62].
EGFR mutations. One study examined prostate cancer patients who Alternatively, IGF signaling can be intercepted by using kinase in-
were unresponsive to gefitinib based therapy and revealed no such hibitors targeting IGFRs. Unfortunately, the tyrosine kinase domain of
sensitizing mutations in any patients [52]. In addition, in vitro studies IGF-IR and the Insulin Receptor (IR) are highly conserved, with a ho-
indicate that a low EGFR/HER2 ratio and PTEN absence are also main mology of 84% [63]. This poses a major challenge to identifying IGF-IR
factors responsible for resistance to erlotinib and gefitinib [53]. De- specific therapeutic inhibitors, as cross-activity against IR would carry
spite the lack of effectiveness of gefitinib as a monotherapy in HRPC, a great risk of diabetogenesis. INSM-18 (nordihydroguaiaretic acid,
combination of gefitinib with radio- or chemo-therapies have demon- NDGA, Fig. 3) is a TKI that selectively inhibits IGF-1R and currently
strated some positive results. Joensuu et al. reported that the combi- in phase II trial for prostate cancer [64]. In an animal model of pros-
nation of gefitinib and radiation has promising activity against tate cancer, INSM-18 demonstrated anti-tumor activity but had no ef-
nonmetastatic prostate cancer in a phase I/II trial [54]. fect on blood glucose levels, indicating that it has minimal off-target
Although the EGFR/HER2 signaling pathways are attractive targets activity for IR. Interestingly, INSM-18 can also inhibit HER2, another
for prostate cancer therapy, there remain several issues that need to kinase that plays a role in prostate cancer. INSM-18 has an IC50 of
be resolved before prostate cancer patients can fully benefit from in- 31 μM for the inhibition of IGF-1 induced autophosphorylation of
hibitors for these targets. The first question relates to the ErbB recep- IGF-IR, and its activity to inhibit ligand-independent tyrosine phos-
tor expression in prostate cancer. It has become clear that tumors phorylation of HER2 is determined to be 15 μM [65]. This characteris-
need to over-express EGFR or HER2 receptors to be effectively tar- tic of INSM-18 renders it more appealing as a therapy for prostate
geted by these inhibitors. However, clinical tests for HER2 and espe- cancer.
cially EGFR (including identification of drug sensitive mutants) need
to be developed to efficiently identify patients for therapies. Secondly,
recent understanding of the effect of downstream molecules, such as 3.3. Platelet-derived growth factor inhibitors
the k-ras mutation, on the resistance to EGFR targeted therapies has
to be incorporated into the screening of prostate patients. This will Platelet-derived growth factors (PDGFs) contain four members of
help to identify patients that are less likely to be benefited from the different polypeptides: PDGF-A, PDGF-B, PDGF-C and PDGF-D [66].
treatment. Meanwhile, simultaneously targeting ErbB receptors and PDGFs can form homodimers or heterodimers via disulfide bonds,
downstream molecules, such as Src [55], might provide an option five of which have so far been described: PDGF-AA, PDGF-AB, PDGF-
for patients who are resistant to the antibody-based therapies. BB, PDGF-CC and PDGF-DD. These factors exert their cellular effects
through receptors PDGFR-α and PDGFR-β. PDGFR-α can be activated
3.2. Inhibitors for insulin-like growth factor (IGF) receptors by PDGF-AA, PDGF-AB, PDGF-BB and PDGF-CC, whilst PDGFR-β is
only bound and activated by PDGF-BB and PDGF-DD. There is an in-
The insulin-like-growth factor (IGF)–IGF receptor (IGF-R) signal- creasing body of evidence implicating PDGFs in the development of
ing pathway plays a crucial role in the cell growth and development solid tumors. Govindarajan demonstrated that SV7tert cells overex-
of both normal and tumor cells [56]. From an evolutionary standpoint, pressing PDGF-BB induce tumor formation when transplanted into
this highly conserved pathway arose to regulate cellular proliferation nude mice [67], documenting that constitutive PDGF signaling can
in response to nutrient availability. The IGF signaling pathway con- act as a critical factor in the malignant transformation of human
sists of two ligands (IGF-I and IGF-II), two cell surface receptors cells. In prostate cancer, overexpression of PDGFRα has been detected
(IGF-IR and IGF-IIR), six specific high affinity binding proteins in epithelial and stromal cells of prostate adenocarcinomas as well
(IGFBP-1 to IGFBP-6), and several other IGFBP-interacting molecules as in bone marrow of metastatic androgen-independent disease,
W. Fu et al. / Biochimica et Biophysica Acta 1825 (2012) 140–152 145
indicating a role for this receptor in both primary and progression of in treating patients with progressive prostate cancer and bone metas-
prostate cancer [68, 69]. tases. Unfortunately, worsened disease progression was observed in
Expression of PDGFR in prostate tumor-associated endothelial patients treated with Tandutinib, although PDGFR inhibition was ob-
cells was much higher than prostate tumor cells [68]. Tumor cells ap- served [78]. It is suspected that PDGF may play multiple roles in pros-
pear to interact with host factors in the microenvironment to induce tate cancer, including the regulation of physiologic equilibrium
growth and the expansion of vasculature. PDGFR inhibitors can, (homeostasis) of bone metastases associated with advanced prostate
therefore, be considered as a class of antivascular therapies that de- cancer. Meanwhile, Tandutinib needs to be improved to have more
stroy tumor cells by blocking the required oxygen and nutrients for selective activity targeting only PDGFR before it can be further tested
survival. We will discuss anti-angiogenesis inhibitors in a later in prostate cancer.
section.
A study by Dolloff et al. found that PC3-ML cells, which possess 4. Anti-angiogenesis targeting therapy
high bone-metastatic potential, express significantly higher levels of
PDGFR compared to malignant but nonmetastatic PC3-N cells [70]. Angiogenesis, the process of new blood vessel formation, is a cru-
When cells were exposed to PDGF, the Akt pathway was activated cial step in the propagation of malignant tumor growth and metasta-
more profoundly in PC3-ML cells. In contrast, there was no difference sis. Tumor growth is highly dependent on diffusion of nutrients and
in EGFR expression and signaling between these two types of cells, wastes, so a steady blood supply is critical to tumor development
suggesting a determinant role for PDGF, but not EGF, in regulating [79]. Among the multiple pro-angiogenic factors that promote the
survival of bone-metastatic prostate cells. The study also postulates process of vessel formation, vascular endothelial growth factor
that the expression of PDGFR can identify cells within the primitive (VEGF) is one of the most important. Clinical trials have demonstrat-
tumor with the highest propensity to metastasize to the skeleton. ed the efficacy of anti-VEGF therapy as a treatment for many types of
Imatinib (Fig. 3) was initially found to block the kinase activity of cancers. The VEGF family of polypeptide growth factors, of which at
the Bcr-Abl fusion oncoprotein that is involved in chronic myeloid least 7 members have been described, activates VEGFR receptor tyro-
leukemia. Later Imatinib was found to be a potent inhibitor of sine kinases resulting in multiple downstream effects [80]. VEGF-A
PDGFR kinase as well [71]. Imatinib showed profound activity for has been shown to associate with key events in tumor angiogenesis.
prostate cancer in animal models. Bone lesions of xenografted mice VEGF-A binds two major receptor VEGF tyrosine receptor kinases,
carrying PC-3MM2-MDR tumors responded to systemic administra- namely VEGFR-1 that is expressed mainly in vascular endothelial
tion of imatinib and paclitaxel (but not to paclitaxel alone), raising cells, and VEGFR-2, which is important in cell trafficking [81, 82].
the possibility that imatinib could have sensitized the tumor cells to The third VEGF receptor, VEGFR3, is primarily associated with lym-
paclitaxel [72]. Meanwhile, after the co-treatment, extensive apopto- phangiogenesis [83].
sis was observed in both tumor cells and tumor associated endotheli- Angiogenesis plays a critical role in prostate cancer and is associat-
al cells [73]. A phase I study of 28 androgen-independent patients ed with higher Gleason grade, metastasis, and worse clinical out-
further confirmed the activity of imatinib in combination with doce- comes. Weidner and colleagues demonstrated that microvessel
taxel [74]. When imatinib was administered alone, only 2 out 28 pa- density (MVD) was significantly higher in prostate cancer samples
tients showed PSA decline (both b50%). After the patients were for those patients with metastatic disease when compared with
treated by the combination of imatinib and docetaxel, decline in those without metastatic disease [84]. A study by Borre et al. in 221
PSA levels were seen in 59% of patients. However, in a follow-up prostate cancer patients followed for a median of 15 years revealed
trial with metastatic CRPC patients, no difference in progression- that MVD of tumor samples at diagnosis was statistically significantly
free survival was observed for the Imatinib plus docetaxel interven- correlated with stage, grade, and disease survival [85]. Furthermore,
tion [75]. Additional investigation is required to determine the activ- serum levels of the ligand VEGF were found to be significantly higher
ity of imatinib for prostate cancer. The unexpected outcomes of the in those prostate cancer patients with metastatic disease [86]. Plasma
clinical trial might be explained by the preferential activity of imati- VEGF levels have also been shown to be an independent prognostic
nib against the osteoclastic response, which is dominant in the PC3- factor in men with metastatic prostate cancer [87, 88]. Based on
MM2 orthotopic model, over the osteoblastic response, which is these findings, angiogenesis has been targeted as a strategy to treat
much more commonly seen in human prostate cancer bone metasta- prostate cancer.
ses [75]. Bevacizumab (Avastin) is a humanized murine monoclonal anti-
SU101 (ARAVA, leflunomide, Fig. 3) is a small organic molecule body against the VEGF receptor and has been shown to have activity
that selectively inhibits PDGFRα and PDGFRβ in vitro [76]. SU101 in multiple cancer cell lines. Bevacizumab is currently FDA-approved
was first developed as a dihydroorotate dehydrogenase inhibitor for the treatment of several malignancies including colorectal carci-
to prevent pyrimidine synthesis. Clinically SU101 exhibits significant noma, NSCLC, recurrent glioblastoma and, most recently, metastatic
anti-inflammatory effects and is currently used as a disease- renal cell carcinoma. Initial preclinical studies showed that VEGF
modulating agent in the treatment of rheumatoid arthritis. In 1997, inhibition by Bevacizumab prevented further tumor growth of the
Shawver and co-workers reported that SU101 mediated inhibition prostate cancer cell line DU145 implanted in nude mice [89]. Unfor-
of mitogenesis induced by PDGF, but not by EGF [76]. In a phase II tunately, use of Bevacizumab as a single agent in prostate cancer
trial with SU101 in hormone refractory prostate cancer, PDGFR ex- was disappointing.
pression was detected in the majority of patients (80% of metastases In a phase II trial, in which 15 patients were treated with 10 mg/kg
and 88% of primary prostate cancers). For patients treated with Bevacizumab every 14 days for six infusions, objective responses or
SU101 as a single agent, 3 out of 39 demonstrated PSA reduction of meaningful PSA level declines were not observed [90]. However, in
more than 50%, one out of 19 patients had partial response for mea- 2010, two phase II clinical trials involving the use of bevacizumab in
surable disease, and 9 out of 35 people reported significant improve- combination with standard chemotherapy had positive results. 60 pa-
ment of pain [77]. SU101 has recently completed a phase II/III trial tients with progressive metastatic CRPC were enrolled and received
with mitixantrone and prednisone for stage IV prostate cancers. intravenous docetaxel and bevacizumab plus oral thalidomide and
Tandutinib (MLN518, Fig. 3), previously known as CT53518, is a prednisone. 90% of the patients had a PSA decline of greater than
small-molecule inhibitor of the type III receptor tyrosine kinases, in- 50%. Progression free survival was estimated to be 18.2 months,
cluding the Fms-like tyrosine kinase 3 receptor (FLT3), platelet- with median overall survival reported as 28.2 months [91].
derived growth factor receptor (PDGFR), and c-Kit receptor tyrosine In another phase II study, 79 patients were enrolled, and 77 of
kinase. Matthew et al. conducted a phase II trial studying Tandutinib those patients were eligible and assessable. Therapy was based on
146 W. Fu et al. / Biochimica et Biophysica Acta 1825 (2012) 140–152
21-day cycles. Patients received oral estramustine phosphate 3 times 5. Other molecular targets for prostate cancer therapy
daily for 5 days, decadron twice daily for 3 days and intravenous doc-
etaxel (70 mg/m 2) and bevacizumab (15 mg/kg). A 50% PSA decline 5.1. Prostate-specific membrane antigen (PSMA)
was observed in 58 patients (75%). Twenty-three of 39 patients
(59%) with measurable disease had a partial response [92]. More re- PSMA is a type II transmembrane glycoprotein predominantly
cently, a randomized phase III trial has been conducted comparing expressed in the prostate epithelium. Significantly increased expres-
the combined use of docetaxel, prednisone, and bevacizumab with sion of PSMA is observed in prostate cancer, particularly in poorly dif-
docetaxel. This study randomized 1050 chemotherapy-naïve meta- ferentiated, metastatic, and hormone refractory carcinomas [107].
static CRPC patients into each treatment group using overall survival The first PSMA mAb, 7E11-C5.3, recognizes intracellular domain of
as a primary endpoint. Despite an improvement in progression-free PSMA. As a result, 7E11-C5.3 (or the antibody conjugate Capromab
survival, disease response and PSA decline, adding bevacizumab to pendetide or CYT-365) only binds to late stage tumors or soft tissue
docetaxel and prednisone did not improve overall survival in patients metastases when cell membrane becomes permeable due to tumor
with metastatic CRPC, and was associated with greater morbidity and necrosis [108].
mortality [93]. An antibody named J591 was developed to target the external do-
Inhibition of tyrosine kinase activity of VEGF receptors is another main of PSMA [146]. Radiolabeled J591 has been shown to accurately
approach to inhibit angiogenesis. Two VEGFR tyrosine kinase inhibi- target bone and soft tissue metastatic prostate cancer sites and may
tors, Sorafenib and Sunitinib (Fig. 3), have been studied for the treat- be useful as therapeutic and diagnostic imaging agents [147]. This an-
ment of prostate cancer. Sorafenib was initially developed to inhibit tibody is currently under evaluation for prostate and non-prostate
Raf-1/BRAF but was subsequently discovered to inhibit some radio-immunotherapies. MLN2704 (Millennium Pharmaceuticals,
receptor tyrosine kinase receptors, including human VEGFR-2, with Inc., Cambridge, MA) is the humanized J591 conjugated with the mi-
IC50 in the nanomolar range [94]. In prostate cancer cells, Sorafenib crotubule-depolymerizing drug maytansinoid 1 (DM1, Immunogen,
has been shown to block the MAPK pathway and induce apoptosis Cambridge, MA) [109]. Phase I/II trials using MLN2704 have revealed
and autophagy [95]. It is expected that Sorafenib, with anti-angiogen- acceptable safety of the compound and the non-immunogenicity of
esis capability, can have more profound in vivo activity. Interestingly, the antibody. In addition, two patients showed sustained PSA decline
in a transgenic mouse adenocarcinoma prostate model, Sorafenib, of more than 50% and six patients had stable PSA levels for up to
mainly through its anti-angiogenesis effect, reduced progression of 86 days [110]. In other trials, huJ591 was radio-labeled with 111indi-
90
high grade prostate intraepithelial neoplasia to adenocarcinoma um (for imaging), yttrium (for therapy), or 177lutetium (for both im-
[96]. In clinical studies, the activity of Sorafenib was initially unclear aging and therapy) [111]. The labeled antibody was shown to target
when PSA was used as the biomarker to measure response. In one bone and/or soft tissue lesions. Kuroda et al. reported the conjugation
open-label phase II study using Sorafenib as the single agent therapy, of saporin toxin with huJ591 and demonstrated anti-tumor activity in
two patients appeared to have worsening PSA scores after treatment animal models [112].
but both had dramatic reduction of bone metastatic lesions as deter-
mined by bone scan [97]. In another phase II trial with CRPC, the PSA 5.2. Heat shock protein (Hsp)
response rate was only 3.6% during the trial, but 10 out of 16 Sorafe-
nib treated patients had PSA declines of 7%–52% after the trial is fin- Hsp is a family of chaperones that assist in the posttranslational
ished [98]. It is suspected that Sorafenib treatment might cause PSA folding of proteins. They play roles in a variety of cellular process in-
release. Thus, response to treatment should be evaluated indepen- cluding proliferation, differentiation, and survival by maintaining the
dent of PSA. A further study concluded that Sorafenib has moderate stability of regulatory proteins [113]. One such Hsp is Hsp90. Inhibi-
activity as a second-line treatment for metastatic CRPC, with one par- tion of Hsp90 function has been shown to cause degradation of client
tial response, 10 stable disease and median survival of 18.3 months proteins via the ubiquitin–proteasome pathway, resulting in the de-
for a total of 46 patients [99]. In comparison, treatment with cytotoxic pletion of multiple cancer-related proteins, including HER2. 17-(ally-
agents in CRPC after docetaxel led to median overall survival in the lamino)-17-demethoxygeldanamycin (17-AAG, Fig. 3), an Hsp90
range of 9.8 months to 17 months. inhibitor derived from geldanamycin, binds to the ATP-binding pock-
Sunitinib is another multi-specific tyrosine kinase inhibitor that et in the N-terminal domain of Hsp90 and blocks the binding of the
targets VEGFR1-3, PDGFRα/β, CSF-1R, and Flt-3 [100]. It is FDA- adenine nucleotides to Hsp90. Interestingly, 17-AAG has much higher
approved for gastrointestinal stromal tumor (as a second line treat- affinity to Hsp90 from tumor than that from normal cells[114]. How-
ment after imatinib), metastatic renal cell carcinoma, and recently ever, in a phase II clinical study 17-AGG did not show any activity in
pancreatic neuroendocrine tumors. In vivo studies showed additive metastatic androgen independent prostate cancer patients with re-
anti-tumor effect of Sunitinib in combination with docetaxel and/or gard to PSA response [115]. F4, a novobiocin analogue designed to in-
anti-EGFR antibody Cetuximab in the prostate cancer PC3 and hibit the C-terminal portion of Hsp90, demonstrated improved
DU145 xenografts [101, 102]. cytotoxicity in prostate cancer cell lines as compared to 17-AAG
In a phase II clinical study, 17 patients with no prior chemotherapy [116]. Recently, a class of small molecule Hsp90 inhibitor gamitrinibs
and 17 patients with docetaxel-resistant prostate cancer were treated showed preclinical activity and favorable safety in the PC3 xenograft
with sunitinib [103]. One patient in each group showed PSA reduction model for drug-resistant and bone metastatic prostate cancer [117].
of more than 50% from the base line. About half of patients in each Gamitrinibs are ATPase antagonists but only accumulate in the mito-
group also reported stable PSA levels at week 12. In addition, improve- chondria of tumor cells, and thus has much less toxicity to normal
ment of radiological imaging was seen in some people even without cells [118].
PSA decline. The activity of sunitinib was also confirmed in other
phase II studies in CRPC [104, 105]. In a very recent phase III trial of suni- 5.3. mTOR
tinib in combination with prednisone as compared to prednisone alone
in metastatic CRPC, treated patients had significant improvement of The phosphoinositide-3 kinase pathway (PI3K) is another critical
progression-free survival, and higher overall response rate were docu- signal transduction pathway that is involved in cancer development.
mented. Overall survival was also longer for sunitinib treated patients This pathway may be activated by growth factors such as EGF, and it
but the difference was not significant (13.1 vs 11.7 months) [106]. Be- leads to the activation of a number of important downstream cellu-
cause of the lack of significant improvement of overall survival in the lar signaling components. One such component is the mammalian
treatment regimen, Pfizer has discontinued the trial. target of rapamycin (mTOR) protein. Activation of mTOR results in
W. Fu et al. / Biochimica et Biophysica Acta 1825 (2012) 140–152 147
sequential activation of downstream molecules, which ultimately several molecules have recently emerged to be potential “targeted”
leads to cell division. Current mTOR Inhibitors in clinical trials are biomarkers that can be used to guide therapies.
mainly rapamycin and temsirolimus (CCI-779) (Fig. 3). The response
of prostate cancer cell lines to a combination of rapamycin and the 6.1. The F77 antigen
nonsteroidal androgen receptor (AR) antagonist bicalutamide has
been documented [119]. In a clinical study involving HRPC patients, Recently, F77, a monoclonal antibody developed by immunizing
two patients treated with Rapamycin experienced a response with mice with prostate cancer cells PC3, was shown to recognize small mo-
PSA reduction, and 4 patients were reported to have stable disease lecular species with glycolipid properties [130]. The F77 antigen,
after the treatment [120]. Wu et al. studied the effects of temsiroli- termed prostate cancer lipid antigen (PCLA), is concentrated in the
mus on hormone resistant PC3 cells, which are PTEN-negative and lipid raft microdomains that serve as platforms in the assembly of asso-
have higher Akt and mTOR activity, and DU145 cells, which are ciating protein complexes. Due to its glycolipid properties, PCLA holds
PTEN positive. Temsirolimus inhibited the growth of xenografts de- significant value as a cancer biomarker. It is expressed on both andro-
rived from both cell lines but better activity was observed with PC3 gen dependent and androgen independent prostate cancer, making it
tumors [121]. a target for early diagnosis and treatment of advanced diseases. Tissue
microarray studies show that F77 stained 112 of 166 primary and 29
5.4. Histone deacetylase (HDAC) of 34 metastatic prostate cancer specimens [130]. By eliciting ADCC/
ACD activity towards the cancer cells, the F77 antibody was useful as a
Epigenetic modifications play a key role in the patho-physiology therapeutic reagent to limit the growth of prostate cancer growth in
of cancer. Histone deacetylases (HDACs), whose substrates are not xenografted mice [130]. Currently the antibody is subjected to human-
limited to histone, are involved in cancer progression. HDACs are ization. The humanized antibody will be tested if F77 provides a novel
part of a transcriptional co-repressor complex that influences various treatment for human prostate cancer.
tumor suppressor genes. Because of the significant roles played by
HDACs in various human cancers, HDAC inhibitors are emerging as 6.2. Serine peptidase inhibitor Kazal type 1 (SPINK1)
a new class of chemotherapeutic agents. HDAC inhibitors have been
shown to induce growth arrest, differentiation, and/or apoptosis of SPINK1 is a biomarker specific to a subset of aggressive prostate can-
prostate cancer cells. Up to now, more than 100 clinical trials are on- cer that does not carry genetic rearrangement of the ETS (E26 transfor-
going with HDAC inhibitors (HDACi) either as monotherapy or in var- mation specific) transcription factors into the TMPRSS2 promoter
ious combination therapies. In one report on a phase II clinical trial region. In 2008, Tomlins et al. demonstrated that high levels of SPINK1
with the HDAC inhibitor romidepsin (Fig. 3), two of 35 HRPC patients in this subset were correlated with a greater rate of cancer recurrence
achieved a confirmed radiological response along with a PSA decline [131]. Recent in vivo data suggests that SPINK1 promotes prostate
of >50% [122]. However, 11 people experienced toxicity and were re- tumor growth through EGFR [132]. SPINK1 has structural similarities
moved from the trial, which raises safety concerns. HDAC inhibitors with EGF and binds to EGFR, inducing the dimerization and sustained
have to be improved for specificity, and combination therapies should phosphorylation of the receptor. Inhibiting SPINK1 attenuates key
be considered. downstream mediators of the EGFR pathway, including MEK, ERK, and
AKT. These results support the potential of EGFR inhibitors to treat
6. New targets and biomarkers for prostate cancer therapy SPINK1 positive prostate cancer. In fact, the anti-EGFR mAb Cetuximab
specifically decreased SPINK1+ tumor growth in mice [132].
One of the validated and widely accepted prostate cancer bio-
markers is PSA. Although the measurement of PSA is clinically prac- 6.3. Survivin
ticed to provide a way to predict response to therapy, this test fails
for some patients. While PSA is elevated in the presence of prostate Survivin is a member of the Inhibitor of Apoptosis (IAP) family that
cancer, it is also present in small amounts in the serum of healthy functions to inhibit caspase activation, leading to the prevention of ap-
males. The effectiveness is thus limited with a positive predictive optosis [133]. As this protein is highly expressed during fetal develop-
value of 28–35% [123]. A recent study also revealed that the change ment and in many human tumors, but also absent in normal
of the PSA levels (PSA velocity) was a poor biomarker for prostate terminally differentiated tissues, it is useful as both a cancer diagnostic
cancer diagnosis and unable to predict the grade (e.g. Gleason marker and therapeutic target [134]. Survivin is overexpressed in mul-
score) of the disease [124]. For some experimental therapies as we tiple types of adenocarcinomas, including prostate [135]. Zhang et al.
mentioned above, reduction of PSA levels is not observed in patients demonstrated Survivin's contribution to the development of hormonal
showing radiological responses. In some cases, the PSA levels can therapy resistance in prostate cancer cells by studying its expression
even rise in patients showing response to therapies by other mea- in LNCaP, an androgen-dependent cell line [136]. It was shown that
surements [97]. inhibiting Survivin could enhance the therapeutic effect of Flutamide,
A great effort in the field of prostate cancer has been made to dis- an anti-androgen agent. However, it was reported that in locally ad-
cover novel prostate markers. This includes extensive effort at the vanced prostate cancer cytoplasmic overexpression of Survivin pre-
proteomic scale [125] and the expansion of biomarker candidates dicted local progression but nuclear expression was associated with
from protein to DNA/RNA [126] and circulating tumor cells [127]. Re- improved survival [137]. Recent studies have revealed that acetylation
views of prostate biomarkers and recent developments are presented promotes the translocation of Survivin into the nucleus, where it func-
in other articles [128, 129]. tions as a repressor of STAT3 oncogenic activity [138]. These data indi-
In many cases, the molecular target for a targeted therapy can be cate that Survivin may have multiple roles in the cell.
used as a biomarker for prostate cancer. Whether or not these bio- Several small-molecule inhibitors and natural compounds that sup-
markers are specific to prostate, their existence in the tumor provides press Survivin expression have been developed and proven effective in
guidance to targeted therapies to obtain higher response in a sub- suppressing prostate cancer tumor growth and enhancing docetaxel-in-
category of prostate cancer. In fact, the unique linkage between the duced apoptosis. One such agent is YM155 (Fig. 3), a small molecule sup-
target and the therapy differentiates targeted therapies from the con- pressant of Survivin expression, promoting apoptosis in vitro and in a
ventional cytotoxic chemotherapeutic therapies for cancers. In addi- xenograft mouse model in vivo [139]. YM155 has completed various
tion to targeted proteins such as the PSMA we mentioned above, phase I and II trials for its safety and efficacy in several types of cancers
148 W. Fu et al. / Biochimica et Biophysica Acta 1825 (2012) 140–152
Table 1
Therapeutic agents and their targets in prostate cancer. mAb: monoclonal antibody; SMI: small molecule inhibitor; TKI: tyrosine kinase inhibitor; doxo: doxorubicin; DCT: doce-
taxel; RTK: receptor tyrosine kinase; PFS: progression free survival; OS: overall survival.
[140, 141]. As a single-agent, YM155 induced decline in PSA concentra- CYP17 17-α-hydroxylase/C17-20-lyase
tions in two out of nine advanced prostate cancer patients [142]. DC dendritic cell
Sun et al. have identified Sanguinarine (Fig. 3), a benzophenanthri- DHEA dehydroepiandrosterone
dine alkaloid primarily derived from the bloodroot plant, as a novel in- EGF epidermal growth factor
hibitor of Survivin [143]. Sanguinarine selectively kills prostate cancer EGFR EGF receptor
cells over “normal” prostate epithelial cells. By promoting Survivin deg- FDA Food and Drug Administration
radation via the ubiquitin–proteasome system, Sanguinarine was found HDAC histone deacetylases
to induce apoptosis and inhibit in vivo tumor formation. HER human EGFR family (HER/ErbB) receptors
Recently, an allosteric modulator S12 (Fig. 3) was developed to bind HRPC hormone refractory prostate cancer
to a cavity near the dimerization domain of Survivin [144]. S12 was able HSP heat shock protein
to change spindle formation and halt mitosis, leading to cell death. It IAP inhibitor of apoptosis
showed anti-tumor activity in several animal models [144]. IGF insulin-like-growth factor
IGF-R IGF receptor
7. Summary IR insulin receptor
LH–RH luteinizing hormone
The rapid expansion of targeted therapies in the past decade has LH–RH luteinizing hormone-releasing hormone
provided new strategies for the treatment of prostate cancer, espe- NSCLC non-small cell lung cancer
cially the most malignant hormone independent and castration resis- PDGF platelet-derived growth factor
tant forms. However, the most advanced therapies in this category PDGFR PDGF receptor
(Table 1), except androgen inhibitors, are still under clinical trials. PSA prostate specific antigen
Further studies will determine the clinical use of those agents that PSMA prostate-specific membrane antigen
show promises in early stage trials. Extensive studies will also be RTK receptor tyrosine kinase
needed to optimize some lead compounds for better specificity and SPINK1 serine peptidase inhibitor Kazal type 1
higher efficacy. Although some of the initial excitements, including VEGF vascular endothelial growth factor
the endothelin A receptor antagonist, zibotentan, failed to be materi-
alized, the extensive research activities in this field have generated
insights for the development of future therapies that could be more
Acknowledgment
efficient to control this disease. In addition to these antibody or
small molecule based approaches, dendritic cell (DC) based therapies
This work was supported by grants from the National Institutes
have also been explored [145], leading to the FDA approved Provenge
of Health (1R01CA157766-01 (A.T.) and R01 CA055306 (M.I.G.))
(sipuleucel-T). The hope is that, with targeted therapies, prostate
and the Abramson Family Cancer Research Institute (to M.I.G.). We
cancer will be divided into sub-categories and treated in a more tar-
thank Dr. Mark I. Greene for his comments on this manuscript.
geted manner in the future.
Abbreviations References
AR androgen receptor
[1] M. Han, A.W. Partin, M. Zahurak, S. Piantadosi, J.I. Epstein, P.C. Walsh, Biochem-
CAPRA Cancer of Prostate Risk Assessment ical (prostate specific antigen) recurrence probability following radical prosta-
CRPC castration resistant prostate cancer tectomy for clinically localized prostate cancer, J. Urol. 169 (2003) 517–523.
W. Fu et al. / Biochimica et Biophysica Acta 1825 (2012) 140–152 149
[2] S.J. Freedland, E.B. Humphreys, L.A. Mangold, M. Eisenberger, F.J. Dorey, P.C. association with high-grade and advanced-stage carcinomas, Mod. Pathol. 23
Walsh, A.W. Partin, Risk of prostate cancer-specific mortality following bio- (2010) 703–712.
chemical recurrence after radical prostatectomy, Jama 294 (2005) 433–439. [25] T. Schlomm, P. Kirstein, L. Iwers, B. Daniel, T. Steuber, J. Walz, F.H. Chun, A.
[3] M.R. Cooperberg, A.J. Vickers, J.M. Broering, P.R. Carroll, Comparative risk- Haese, J. Kollermann, M. Graefen, H. Huland, G. Sauter, R. Simon, A. Erbersdobler,
adjusted mortality outcomes after primary surgery, radiotherapy, or Clinical significance of epidermal growth factor receptor protein overexpression
androgen-deprivation therapy for localized prostate cancer, Cancer 116 (2010) and gene copy number gains in prostate cancer, Clin. Cancer Res. 13 (2007)
5226–5234. 6579–6584.
[4] A.J. Stephenson, M.W. Kattan, J.A. Eastham, Z.A. Dotan, F.J. Bianco Jr., H. Lilja, P.T. [26] B. Shuch, M. Mikhail, J. Satagopan, P. Lee, H. Yee, C. Chang, C. Cordon-Cardo, S.S.
Scardino, Defining biochemical recurrence of prostate cancer after radical pros- Taneja, I. Osman, Racial disparity of epidermal growth factor receptor expres-
tatectomy: a proposal for a standardized definition, J. Clin. Oncol. 24 (2006) sion in prostate cancer, J. Clin. Oncol. 22 (2004) 4725–4729.
3973–3978. [27] A.S. Neto, M. Tobias-Machado, M.L. Wroclawski, F.L. Fonseca, G.K. Teixeira, R.D.
[5] J.S. de Bono, S. Oudard, M. Ozguroglu, S. Hansen, J.P. Machiels, I. Kocak, G. Gravis, Amarante, E.R. Wroclawski, A. Del Giglio, Her-2/neu expression in prostate ade-
I. Bodrogi, M.J. Mackenzie, L. Shen, M. Roessner, S. Gupta, A.O. Sartor, Prednisone nocarcinoma: a systematic review and meta-analysis, J. Urol. 184 (2010)
plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate 842–850.
cancer progressing after docetaxel treatment: a randomised open-label trial, [28] Y. Liu, S. Majumder, W. McCall, C.I. Sartor, J.L. Mohler, C.W. Gregory, H.S. Earp,
Lancet 376 (2010) 1147–1154. Y.E. Whang, Inhibition of HER-2/neu kinase impairs androgen receptor recruit-
[6] C.A. Heinlein, C. Chang, Androgen receptor in prostate cancer, Endocr. Rev. 25 ment to the androgen responsive enhancer, Cancer Res. 65 (2005) 3404–3409.
(2004) 276–308. [29] T. Karashima, P. Sweeney, J.W. Slaton, S.J. Kim, D. Kedar, J.I. Izawa, Z. Fan, C.
[7] A.F. Santos, H. Huang, D.J. Tindall, The androgen receptor: a potential target for Pettaway, D.J. Hicklin, T. Shuin, C.P. Dinney, Inhibition of angiogenesis by the
therapy of prostate cancer, Steroids 69 (2004) 79–85. antiepidermal growth factor receptor antibody ImClone C225 in androgen-
[8] I.U. Agoulnik, N.L. Weigel, Androgen receptor action in hormone-dependent and independent prostate cancer growing orthotopically in nude mice, Clin. Cancer
recurrent prostate cancer, J. Cell. Biochem. 99 (2006) 362–372. Res. 8 (2002) 1253–1264.
[9] J.A. Locke, E.S. Guns, A.A. Lubik, H.H. Adomat, S.C. Hendy, C.A. Wood, S.L. Ettin- [30] P. Dhupkar, M. Dowling, K. Cengel, B. Chen, Effects of anti-EGFR antibody cetux-
ger, M.E. Gleave, C.C. Nelson, Androgen levels increase by intratumoral de imab on androgen-independent prostate cancer cells, Anticancer. Res. 30 (2010)
novo steroidogenesis during progression of castration-resistant prostate cancer, 1905–1910.
Cancer Res. 68 (2008) 6407–6415. [31] X.D. Yang, X.C. Jia, J.R. Corvalan, P. Wang, C.G. Davis, Development of ABX-EGF, a
[10] Y. Wu, J.E. Rosenberg, M.E. Taplin, Novel agents and new therapeutics in fully human anti-EGF receptor monoclonal antibody, for cancer therapy, Crit.
castration-resistant prostate cancer, Curr. Opin. Oncol. 23 (2011) 290–296. Rev. Oncol. Hematol. 38 (2001) 17–23.
[11] M.E. Jung, S. Ouk, D. Yoo, C.L. Sawyers, C. Chen, C. Tran, J. Wongvipat, Structure– [32] L.M. Weiner, A.S. Belldegrun, J. Crawford, A.W. Tolcher, P. Lockbaum, R.H.
activity relationship for thiohydantoin androgen receptor antagonists for castration- Arends, L. Navale, R.G. Amado, G. Schwab, R.A. Figlin, Dose and schedule study
resistant prostate cancer (CRPC), J. Med. Chem. 53 (2010) 2779–2796. of panitumumab monotherapy in patients with advanced solid malignancies,
[12] C. Tran, S. Ouk, N.J. Clegg, Y. Chen, P.A. Watson, V. Arora, J. Wongvipat, P.M. Clin. Cancer Res. 14 (2008) 502–508.
Smith-Jones, D. Yoo, A. Kwon, T. Wasielewska, D. Welsbie, C.D. Chen, C.S. Higano, [33] M. Wagener, X. Zhang, H.G. Villarreal, L. Levy, P. Allen, S. Shentu, B. Fang, S.
T.M. Beer, D.T. Hung, H.I. Scher, M.E. Jung, C.L. Sawyers, Development of a Krishnan, J.Y. Chang, M.R. Cheung, Effect of combining anti-epidermal growth
second-generation antiandrogen for treatment of advanced prostate cancer, factor receptor antibody C225 and radiation on DU145 prostate cancer, Oncol.
Science 324 (2009) 787–790. Rep. 19 (2008) 1071–1077.
[13] H.I. Scher, T.M. Beer, C.S. Higano, A. Anand, M.E. Taplin, E. Efstathiou, D. [34] S.F. Slovin, W.K. Kelly, A. Wilton, M. Kattan, P. Myskowski, J. Mendelsohn, H.I.
Rathkopf, J. Shelkey, E.Y. Yu, J. Alumkal, D. Hung, M. Hirmand, L. Seely, M.J. Scher, Anti-epidermal growth factor receptor monoclonal antibody cetuximab
Morris, D.C. Danila, J. Humm, S. Larson, M. Fleisher, C.L. Sawyers, Antitumour activ- plus Doxorubicin in the treatment of metastatic castration-resistant prostate
ity of MDV3100 in castration-resistant prostate cancer: a phase 1–2 study, Lancet cancer, Clin. Genitourin. Cancer 7 (2009) E77–E82.
375 (2010) 1437–1446. [35] S. Signoretti, R. Montironi, J. Manola, A. Altimari, C. Tam, G. Bubley, S. Balk, G.
[14] S. Haidar, P.B. Ehmer, S. Barassin, C. Batzl-Hartmann, R.W. Hartmann, Effects Thomas, I. Kaplan, L. Hlatky, P. Hahnfeldt, P. Kantoff, M. Loda, Her-2-neu expres-
of novel 17alpha-hydroxylase/C17, 20-lyase (P450 17, CYP 17) inhibitors on an- sion and progression toward androgen independence in human prostate cancer,
drogen biosynthesis in vitro and in vivo, J. Steroid Biochem. Mol. Biol. 84 (2003) J. Natl. Cancer Inst. 92 (2000) 1918–1925.
555–562. [36] D.B. Agus, H.I. Scher, B. Higgins, W.D. Fox, G. Heller, M. Fazzari, C. Cordon-Cardo,
[15] S.E. Barrie, G.A. Potter, P.M. Goddard, B.P. Haynes, M. Dowsett, M. Jarman, Phar- D.W. Golde, Response of prostate cancer to anti-Her-2/neu antibody in androgen-
macology of novel steroidal inhibitors of cytochrome P450(17) alpha (17 alpha- dependent and -independent human xenograft models, Cancer Res. 59 (1999)
hydroxylase/C17-20 lyase), J. Steroid Biochem. Mol. Biol. 50 (1994) 267–273. 4761–4764.
[16] G.A. Potter, S.E. Barrie, M. Jarman, M.G. Rowlands, Novel steroidal inhibitors of [37] M.E. Legrier, S. Oudard, J.G. Judde, C. Guyader, G. de Pinieux, K. Boye, P. de Cremoux,
human cytochrome P45017 alpha (17 alpha-hydroxylase-C17,20-lyase): poten- B. Dutrillaux, M.F. Poupon, Potentiation of antitumour activity of docetaxel by
tial agents for the treatment of prostatic cancer, J. Med. Chem. 38 (1995) combination with trastuzumab in a human prostate cancer xenograft model
2463–2471. and underlying mechanisms, Br. J. Cancer 96 (2007) 269–276.
[17] C.J. Ryan, M.R. Smith, L. Fong, J.E. Rosenberg, P. Kantoff, F. Raynaud, V. Martins, G. [38] E.J. Small, R. Bok, D.M. Reese, D. Sudilovsky, M. Frohlich, Docetaxel, estramus-
Lee, T. Kheoh, J. Kim, A. Molina, E.J. Small, Phase I clinical trial of the CYP17 in- tine, plus trastuzumab in patients with metastatic androgen-independent pros-
hibitor abiraterone acetate demonstrating clinical activity in patients with tate cancer, Semin. Oncol. 28 (2001) 71–76.
castration-resistant prostate cancer who received prior ketoconazole therapy, [39] H. Zhang, A. Berezov, Q. Wang, G. Zhang, J. Drebin, R. Murali, M.I. Greene, ErbB
J. Clin. Oncol. 28 (2010) 1481–1488. receptors: from oncogenes to targeted cancer therapies, J. Clin. Invest. 117
[18] G. Attard, A.H. Reid, T.A. Yap, F. Raynaud, M. Dowsett, S. Settatree, M. Barrett, C. (2007) 2051–2058.
Parker, V. Martins, E. Folkerd, J. Clark, C.S. Cooper, S.B. Kaye, D. Dearnaley, G. Lee, [40] L. Bonaccorsi, S. Marchiani, M. Muratori, G. Forti, E. Baldi, Gefitinib (‘IRESSA’,
J.S. de Bono, Phase I clinical trial of a selective inhibitor of CYP17, abiraterone ac- ZD1839) inhibits EGF-induced invasion in prostate cancer cells by suppressing
etate, confirms that castration-resistant prostate cancer commonly remains hor- PI3 K/AKT activation, J. Cancer Res. Clin. Oncol. 130 (2004) 604–614.
mone driven, J. Clin. Oncol. 26 (2008) 4563–4571. [41] C. Vicentini, C. Festuccia, G.L. Gravina, A. Angelucci, A. Marronaro, M. Bologna,
[19] D.C. Danila, M.J. Morris, J.S. de Bono, C.J. Ryan, S.R. Denmeade, M.R. Smith, M.E. Prostate cancer cell proliferation is strongly reduced by the epidermal growth
Taplin, G.J. Bubley, T. Kheoh, C. Haqq, A. Molina, A. Anand, M. Koscuiszka, S.M. factor receptor tyrosine kinase inhibitor ZD1839 in vitro on human cell lines
Larson, L.H. Schwartz, M. Fleisher, H.I. Scher, Phase II multicenter study of abir- and primary cultures, J. Cancer Res. Clin. Oncol. 129 (2003) 165–174.
aterone acetate plus prednisone therapy in patients with docetaxel-treated [42] A. Sgambato, A. Camerini, B. Faraglia, R. Ardito, G. Bianchino, D. Spada, A.
castration-resistant prostate cancer, J. Clin. Oncol. 28 (2010) 1496–1501. Boninsegna, V. Valentini, A. Cittadini, Targeted inhibition of the epidermal
[20] C.J. Ryan, S. Shah, E. Efstathiou, M.R. Smith, M.E. Taplin, G.J. Bubley, C.J. Logothetis, growth factor receptor-tyrosine kinase by ZD1839 (‘Iressa’) induces cell-cycle
T. Kheoh, C. Kilian, C.M. Haqq, A. Molina, E.J. Small, Phase II study of abiraterone arrest and inhibits proliferation in prostate cancer cells, J. Cell. Physiol. 201
acetate in chemotherapy-naive metastatic castration-resistant prostate cancer (2004) 97–105.
displaying bone flare discordant with serologic response, Clin. Cancer Res. 17 [43] C. Pezaro, M.A. Rosenthal, H. Gurney, I.D. Davis, C. Underhill, M.J. Boyer, D. Kotasek,
(2011) 4854–4861. B. Solomon, G.C. Toner, An open-label, single-arm phase two trial of gefitinib in
[21] S.K. Pal, O. Sartor, Phase III data for abiraterone in an evolving landscape for patients with advanced or metastatic castration-resistant prostate cancer, Am.
castration-resistant prostate cancer, Maturitas 68 (2011) 103–105. J. Clin. Oncol. 32 (2009) 338–341.
[22] J.S. de Bono, C.J. Logothetis, A. Molina, K. Fizazi, S. North, L. Chu, K.N. Chi, R.J. [44] C.M. Canil, M.J. Moore, E. Winquist, T. Baetz, M. Pollak, K.N. Chi, S. Berry, D.S.
Jones, O.B. Goodman Jr., F. Saad, J.N. Staffurth, P. Mainwaring, S. Harland, Ernst, L. Douglas, M. Brundage, B. Fisher, A. McKenna, L. Seymour, Randomized
T.W. Flaig, T.E. Hutson, T. Cheng, H. Patterson, J.D. Hainsworth, C.J. Ryan, C.N. phase II study of two doses of gefitinib in hormone-refractory prostate cancer:
Sternberg, S.L. Ellard, A. Flechon, M. Saleh, M. Scholz, E. Efstathiou, A. Zivi, D. a trial of the National Cancer Institute of Canada-Clinical Trials Group, J. Clin.
Bianchini, Y. Loriot, N. Chieffo, T. Kheoh, C.M. Haqq, H.I. Scher, Abiraterone Oncol. 23 (2005) 455–460.
and increased survival in metastatic prostate cancer, N. Engl. J. Med. 364 (2011) [45] E.J. Small, J. Fontana, N. Tannir, R.S. DiPaola, G. Wilding, M. Rubin, R.B. Iacona, F.F.
1995–2005. Kabbinavar, A phase II trial of gefitinib in patients with non-metastatic
[23] G. Blackledge, Growth factor receptor tyrosine kinase inhibitors; clinical devel- hormone-refractory prostate cancer, BJU Int. 100 (2007) 765–769.
opment and potential for prostate cancer therapy, J. Urol. 170 (2003) S77–83 [46] C. Nabhan, T.M. Lestingi, A. Galvez, K. Tolzien, S.K. Kelby, D. Tsarwhas, S. Newman,
discussion S83. J.D. Bitran, Erlotinib has moderate single-agent activity in chemotherapy-naive
[24] S. de Muga, S. Hernandez, L. Agell, M. Salido, N. Juanpere, M. Lorenzo, J.A. Lorente, castration-resistant prostate cancer: final results of a phase II trial, Urology 74
S. Serrano, J. Lloreta, Molecular alterations of EGFR and PTEN in prostate cancer: (2009) 665–671.
150 W. Fu et al. / Biochimica et Biophysica Acta 1825 (2012) 140–152
[47] G. Gravis, F. Bladou, N. Salem, A. Goncalves, B. Esterni, J. Walz, S. Bagattini, [69] A. Chott, Z. Sun, D. Morganstern, J. Pan, T. Li, M. Susani, I. Mosberger, M.P. Upton,
M. Marcy, S. Brunelle, P. Viens, Results from a monocentric phase II trial of G.J. Bubley, S.P. Balk, Tyrosine kinases expressed in vivo by human prostate can-
erlotinib in patients with metastatic prostate cancer, Ann. Oncol. 19 (2008) cer bone marrow metastases and loss of the type 1 insulin-like growth factor re-
1624–1628. ceptor, Am. J. Pathol. 155 (1999) 1271–1279.
[48] Y.E. Whang, A.J. Armstrong, W.K. Rathmell, P.A. Godley, W.Y. Kim, R.S. Pruthi, [70] N.G. Dolloff, S.S. Shulby, A.V. Nelson, M.E. Stearns, G.J. Johannes, J.D. Thomas, O.
E.M. Wallen, J.M. Crane, D.T. Moore, G. Grigson, K. Morris, C.P. Watkins, D.J. Meucci, A. Fatatis, Bone-metastatic potential of human prostate cancer cells cor-
George, A phase II study of lapatinib, a dual EGFR and HER-2 tyrosine kinase relates with Akt/PKB activation by alpha platelet-derived growth factor recep-
inhibitor, in patients with castration-resistant prostate cancer, Urol. Oncol. tor, Oncogene 24 (2005) 6848–6854.
(2011) (Electronic publication ahead of print), doi:10.1016/j.urolonc.2010. [71] D. George, Targeting PDGF receptors in cancer—rationales and proof of concept
09.018. clinical trials, Adv. Exp. Med. Biol. 532 (2003) 141–151.
[49] S.S. Sridhar, S.J. Hotte, J.L. Chin, G.R. Hudes, R. Gregg, J. Trachtenberg, L. Wang, D. [72] H. Uehara, S.J. Kim, T. Karashima, D.L. Shepherd, D. Fan, R. Tsan, J.J. Killion, C.
Tran-Thanh, N.A. Pham, M.S. Tsao, D. Hedley, J.E. Dancey, M.J. Moore, A multi- Logothetis, P. Mathew, I.J. Fidler, Effects of blocking platelet-derived growth
center phase II clinical trial of lapatinib (GW572016) in hormonally untreated factor-receptor signaling in a mouse model of experimental prostate cancer
advanced prostate cancer, Am. J. Clin. Oncol. 33 (2010) 609–613. bone metastases, J. Natl. Cancer Inst. 95 (2003) 458–470.
[50] G. Liu, Y.H. Chen, J. Kolesar, W. Huang, R. Dipaola, M. Pins, M. Carducci, M. Stein, [73] S.J. Kim, H. Uehara, S. Yazici, J.E. Busby, T. Nakamura, J. He, M. Maya, C. Logothe-
G.J. Bubley, G. Wilding, Eastern Cooperative Oncology Group Phase II Trial of tis, P. Mathew, X. Wang, K.A. Do, D. Fan, I.J. Fidler, Targeting platelet-derived
lapatinib in men with biochemically relapsed, androgen dependent prostate growth factor receptor on endothelial cells of multidrug-resistant prostate can-
cancer, Urol. Oncol. (2011) (Electronic publication ahead of print), doi:10.1016/ cer, J. Natl. Cancer Inst. 98 (2006) 783–793.
j.urolonc.2011.01.002. [74] P. Mathew, P.F. Thall, D. Jones, C. Perez, C. Bucana, P. Troncoso, S.J. Kim, I.J. Fidler,
[51] J.G. Paez, P.A. Janne, J.C. Lee, S. Tracy, H. Greulich, S. Gabriel, P. Herman, F.J. Kaye, C. Logothetis, Platelet-derived growth factor receptor inhibitor imatinib mesy-
N. Lindeman, T.J. Boggon, K. Naoki, H. Sasaki, Y. Fujii, M.J. Eck, W.R. Sellers, B.E. late and docetaxel: a modular phase I trial in androgen-independent prostate
Johnson, M. Meyerson, EGFR mutations in lung cancer: correlation with clinical cancer, J. Clin. Oncol. 22 (2004) 3323–3329.
response to gefitinib therapy, Science 304 (2004) 1497–1500. [75] P. Mathew, P.F. Thall, C.D. Bucana, W.K. Oh, M.J. Morris, D.M. Jones, M.M. Johnson,
[52] G. Curigliano, G. Pelosi, T. De Pas, G. Renne, O. De Cobelli, M. Manzotti, G. Spitaleri, S. Wen, L.C. Pagliaro, N.M. Tannir, S.M. Tu, A.A. Meluch, L. Smith, L. Cohen, S.J. Kim,
F. de Braud, Absence of epidermal growth factor receptor gene mutations in P. Troncoso, I.J. Fidler, C.J. Logothetis, Platelet-derived growth factor receptor
patients with hormone refractory prostate cancer not responding to gefitinib, inhibition and chemotherapy for castration-resistant prostate cancer with bone
Prostate 67 (2007) 603–604. metastases, Clin. Cancer Res. 13 (2007) 5816–5824.
[53] C. Festuccia, G.L. Gravina, L. Biordi, S. D'Ascenzo, V. Dolo, C. Ficorella, E. Ricevuto, [76] L.K. Shawver, D.P. Schwartz, E. Mann, H. Chen, J. Tsai, L. Chu, L. Taylorson, M.
V. Tombolini, Effects of EGFR tyrosine kinase inhibitor erlotinib in prostate cancer Longhi, S. Meredith, L. Germain, J.S. Jacobs, C. Tang, A. Ullrich, M.E. Berens,
cells in vitro, Prostate 69 (2009) 1529–1537. E. Hersh, G. McMahon, K.P. Hirth, T.J. Powell, Inhibition of platelet-derived
[54] G. Joensuu, T. Joensuu, P. Nokisalmi, C. Reddy, J. Isola, M. Ruutu, M. Kouri, P.A. growth factor-mediated signal transduction and tumor growth by N-[4-
Kupelian, J. Collan, S. Pesonen, A. Hemminki, A phase I/II trial of gefitinib given (trifluoromethyl)-phenyl]5-methylisoxazole-4-carboxamide, Clin. Cancer Res.
concurrently with radiotherapy in patients with nonmetastatic prostate cancer, 3 (1997) 1167–1177.
Int. J. Radiat. Oncol. Biol. Phys. 78 (2010) 42–49. [77] Y.J. Ko, E.J. Small, F. Kabbinavar, A. Chachoua, S. Taneja, D. Reese, A. DePaoli, A.
[55] S. Zhang, W.C. Huang, P. Li, H. Guo, S.B. Poh, S.W. Brady, Y. Xiong, L.M. Tseng, S.H. Hannah, S.P. Balk, G.J. Bubley, A multi-institutional phase ii study of SU101, a
Li, Z. Ding, A.A. Sahin, F.J. Esteva, G.N. Hortobagyi, D. Yu, Combating trastuzumab platelet-derived growth factor receptor inhibitor, for patients with hormone-
resistance by targeting SRC, a common node downstream of multiple resistance refractory prostate cancer, Clin. Cancer Res. 7 (2001) 800–805.
pathways, Nat. Med. 17 (2011) 461–469. [78] P. Mathew, N. Tannir, S.M. Tu, S. Wen, C.C. Guo, V. Marcott, B.N. Bekele, L.
[56] E.K. Rowinsky, H. Youssoufian, J.R. Tonra, P. Solomon, D. Burtrum, D.L. Ludwig, Pagliaro, Accelerated disease progression in prostate cancer and bone metasta-
IMC-A12, a human IgG1 monoclonal antibody to the insulin-like growth factor ses with platelet-derived growth factor receptor inhibition: observations with
I receptor, Clin. Cancer Res. 13 (2007) 5549s–5555s. tandutinib, Cancer Chemother. Pharmacol. 68 (2011) 889–896.
[57] S. Monti, L. Proietti-Pannunzi, A. Sciarra, F. Lolli, P. Falasca, M. Poggi, F.S. Celi, [79] C. Hwang, E.I. Heath, Angiogenesis inhibitors in the treatment of prostate cancer,
V. Toscano, The IGF axis in prostate cancer, Curr. Pharm. Des. 13 (2007) J. Hematol. Oncol. 3 (2010) 26.
719–727. [80] R.J. Epstein, VEGF signaling inhibitors: more pro-apoptotic than anti-angiogenic,
[58] S. Kojima, M. Inahara, H. Suzuki, T. Ichikawa, Y. Furuya, Implications of insulin- Cancer Metastasis Rev. 26 (2007) 443–452.
like growth factor-I for prostate cancer therapies, Int. J. Urol. 16 (2009) 161–167. [81] Q. Zhao, K. Egashira, K. Hiasa, M. Ishibashi, S. Inoue, K. Ohtani, C. Tan, M. Shibuya,
[59] M.R. Cardillo, S. Monti, F. Di Silverio, V. Gentile, F. Sciarra, V. Toscano, Insulin-like A. Takeshita, K. Sunagawa, Essential role of vascular endothelial growth factor
growth factor (IGF)-I, IGF-II and IGF type I receptor (IGFR-I) expression in pros- and Flt-1 signals in neointimal formation after periadventitial injury, Arterios-
tatic cancer, Anticancer. Res. 23 (2003) 3825–3835. cler. Thromb. Vasc. Biol. 24 (2004) 2284–2289.
[60] C.S. Higano, J.J. Alumkal, C.J. Ryan, E.Y. Yu, T.M. Beer, F.E. Fox, A. Dontabhaktuni, [82] H. Zhong, J.P. Bowen, Molecular design and clinical development of VEGFR
H. Youssoufian, J.D. Schwartz, A phase II study of cixutumumab (IMC-A12), a kinase inhibitors, Curr. Top Med. Chem. 7 (2007) 1379–1393.
monoclonal antibody (MAb) against the insulin-like growth factor 1 receptor [83] K. Paavonen, P. Puolakkainen, L. Jussila, T. Jahkola, K. Alitalo, Vascular endothe-
(IGF-IR), monotherapy in metastatic castration-resistant prostate cancer lial growth factor receptor-3 in lymphangiogenesis in wound healing, Am. J.
(mCRPC), 2010 Genitourinary Cancers Symposium, 2010, Abstract # 189. Pathol. 156 (2000) 1499–1504.
[61] D. Burtrum, Z. Zhu, D. Lu, D.M. Anderson, M. Prewett, D.S. Pereira, R. Bassi, R. [84] N. Weidner, P.R. Carroll, J. Flax, W. Blumenfeld, J. Folkman, Tumor angiogenesis
Abdullah, A.T. Hooper, H. Koo, X. Jimenez, D. Johnson, R. Apblett, P. Kussie, P. correlates with metastasis in invasive prostate carcinoma, Am. J. Pathol. 143
Bohlen, L. Witte, D.J. Hicklin, D.L. Ludwig, A fully human monoclonal anti- (1993) 401–409.
body to the insulin-like growth factor I receptor blocks ligand-dependent [85] M. Borre, B.V. Offersen, B. Nerstrom, J. Overgaard, Microvessel density predicts
signaling and inhibits human tumor growth in vivo, Cancer Res. 63 (2003) survival in prostate cancer patients subjected to watchful waiting, Br. J. Cancer
8912–8921. 78 (1998) 940–944.
[62] J.D. Wu, K. Haugk, I. Coleman, L. Woodke, R. Vessella, P. Nelson, R.B. Montgom- [86] J.L. Duque, K.R. Loughlin, R.M. Adam, P.W. Kantoff, D. Zurakowski, M.R. Freeman,
ery, D.L. Ludwig, S.R. Plymate, Combined in vivo effect of A12, a type 1 insulin- Plasma levels of vascular endothelial growth factor are increased in patients
like growth factor receptor antibody, and docetaxel against prostate cancer tumors, with metastatic prostate cancer, Urology 54 (1999) 523–527.
Clin. Cancer Res. 12 (2006) 6153–6160. [87] D.J. George, S. Halabi, T.F. Shepard, N.J. Vogelzang, D.F. Hayes, E.J. Small,
[63] A. Ullrich, A. Gray, A.W. Tam, T. Yang-Feng, M. Tsubokawa, C. Collins, W. Henzel, P.W. Kantoff, Prognostic significance of plasma vascular endothelial growth
T. Le Bon, S. Kathuria, E. Chen, et al., Insulin-like growth factor I receptor primary factor levels in patients with hormone-refractory prostate cancer treated
structure: comparison with insulin receptor suggests structural determinants on Cancer and Leukemia Group B 9480, Clin. Cancer Res. 7 (2001)
that define functional specificity, EMBO J. 5 (1986) 2503–2512. 1932–1936.
[64] Y. Wang, Q.S. Ji, M. Mulvihill, J.A. Pachter, Inhibition of the IGF-I receptor for [88] Y.M. El-Gohary, J.F. Silverman, P.R. Olson, Y.L. Liu, J.K. Cohen, R. Miller, R.S. Saad,
treatment of cancer. Kinase inhibitors and monoclonal antibodies as alternative Endoglin (CD105) and vascular endothelial growth factor as prognostic markers
approaches, Recent Results Cancer Res. 172 (2007) 59–76. in prostatic adenocarcinoma, Am. J. Clin. Pathol. 127 (2007) 572–579.
[65] J.F. Youngren, K. Gable, C. Penaranda, B.A. Maddux, M. Zavodovskaya, M. Lobo, [89] P. Borgstrom, M.A. Bourdon, K.J. Hillan, P. Sriramarao, N. Ferrara, Neutralizing
M. Campbell, J. Kerner, I.D. Goldfine, Nordihydroguaiaretic acid (NDGA) inhibits anti-vascular endothelial growth factor antibody completely inhibits angiogen-
the IGF-1 and c-erbB2/HER2/neu receptors and suppresses growth in breast esis and growth of human prostate carcinoma micro tumors in vivo, Prostate 35
cancer cells, Breast Cancer Res. Treat. 94 (2005) 37–46. (1998) 1–10.
[66] L. Fredriksson, H. Li, U. Eriksson, The PDGF family: four gene products form five [90] David M. Reese, P. Fratesi, M. Corry, W. Novotny, E. Holmgren, Eric J. Small, A
dimeric isoforms, Cytokine Growth Factor Rev. 15 (2004) 197–204. phase II trial of humanized anti-vascular endothelial growth factor antibody
[67] B. Govindarajan, A. Shah, C. Cohen, R.S. Arnold, J. Schechner, J. Chung, A.M. for the treatment of androgen-independent prostate cancer, Prostate J. 3
Mercurio, R. Alani, B. Ryu, C.Y. Fan, J.M. Cuezva, M. Martinez, J.L. Arbiser, Malignant (2001) 65–70.
transformation of human cells by constitutive expression of platelet-derived [91] Y.M. Ning, J.L. Gulley, P.M. Arlen, S. Woo, S.M. Steinberg, J.J. Wright, H.L. Parnes,
growth factor-BB, J. Biol. Chem. 280 (2005) 13936–13943. J.B. Trepel, M.J. Lee, Y.S. Kim, H. Sun, R.A. Madan, L. Latham, E. Jones, C.C. Chen,
[68] K. Fudge, C.Y. Wang, M.E. Stearns, Immunohistochemistry analysis of W.D. Figg, W.L. Dahut, Phase II trial of bevacizumab, thalidomide, docetaxel,
platelet-derived growth factor A and B chains and platelet-derived growth and prednisone in patients with metastatic castration-resistant prostate cancer,
factor alpha and beta receptor expression in benign prostatic hyperplasias J. Clin. Oncol. 28 (2010) 2070–2076.
and Gleason-graded human prostate adenocarcinomas, Mod. Pathol. 7 [92] J. Picus, S. Halabi, W.K. Kelly, N.J. Vogelzang, Y.E. Whang, E.B. Kaplan, W.M.
(1994) 549–554. Stadler, E.J. Small, A phase 2 study of estramustine, docetaxel, and bevacizumab
W. Fu et al. / Biochimica et Biophysica Acta 1825 (2012) 140–152 151
in men with castrate-resistant prostate cancer: results from Cancer and Leukemia A phase II trial of 17-allylamino-17-demethoxygeldanamycin in patients with
Group B Study 90006, Cancer 117 (2011) 526–533. hormone-refractory metastatic prostate cancer, Clin. Cancer Res. 14 (2008)
[93] D. Pouessel, S. Culine, Actualities in prostate cancer in ASCO annual meeting 7940–7946.
2010, Bull. Cancer 97 (2010) 1563–1572. [116] S.B. Matthews, G.A. Vielhauer, C.A. Manthe, V.K. Chaguturu, K. Szabla, R.L. Matts,
[94] S.M. Wilhelm, C. Carter, L. Tang, D. Wilkie, A. McNabola, H. Rong, C. Chen, X. A.C. Donnelly, B.S. Blagg, J.M. Holzbeierlein, Characterization of a novel novobi-
Zhang, P. Vincent, M. McHugh, Y. Cao, J. Shujath, S. Gawlak, D. Eveleigh, B. ocin analogue as a putative C-terminal inhibitor of heat shock protein 90 in
Rowley, L. Liu, L. Adnane, M. Lynch, D. Auclair, I. Taylor, R. Gedrich, A. Voznesensky, prostate cancer cells, Prostate 70 (2010) 27–36.
B. Riedl, L.E. Post, G. Bollag, P.A. Trail, BAY 43-9006 exhibits broad spectrum oral [117] B.H. Kang, M.D. Siegelin, J. Plescia, C.M. Raskett, D.S. Garlick, T. Dohi, J.B. Lian, G.S.
antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine Stein, L.R. Languino, D.C. Altieri, Preclinical characterization of mitochondria-
kinases involved in tumor progression and angiogenesis, Cancer Res. 64 (2004) targeted small molecule hsp90 inhibitors, gamitrinibs, in advanced prostate
7099–7109. cancer, Clin. Cancer Res. 16 (2010) 4779–4788.
[95] A. Ullen, M. Farnebo, L. Thyrell, S. Mahmoudi, P. Kharaziha, L. Lennartsson, D. [118] B.H. Kang, J. Plescia, H.Y. Song, M. Meli, G. Colombo, K. Beebe, B. Scroggins, L.
Grander, T. Panaretakis, S. Nilsson, Sorafenib induces apoptosis and autophagy Neckers, D.C. Altieri, Combinatorial drug design targeting multiple cancer sig-
in prostate cancer cells in vitro, Int. J. Oncol. 37 (2010) 15–20. naling networks controlled by mitochondrial Hsp90, J. Clin. Invest. 119 (2009)
[96] A.V. Bono, T. Pannellini, M. Liberatore, R. Montironi, S.C. Cunico, L. Cheng, F. 454–464.
Sasso, P. Musiani, M. Iezzi, Sorafenib's inhibition of prostate cancer growth in [119] Y. Wang, M. Mikhailova, S. Bose, C.X. Pan, R.W. deVere White, P.M. Ghosh, Reg-
transgenic adenocarcinoma mouse prostate mice and its differential effects on ulation of androgen receptor transcriptional activity by rapamycin in prostate
endothelial and pericyte growth during tumor angiogenesis, Anal. Quant. cancer cell proliferation and survival, Oncogene 27 (2008) 7106–7117.
Cytol. Histol. 32 (2010) 136–145. [120] R.J. Amato, J. Jac, T. Mohammad, S. Saxena, Pilot study of rapamycin in patients
[97] W.L. Dahut, C. Scripture, E. Posadas, L. Jain, J.L. Gulley, P.M. Arlen, J.J. Wright, Y. with hormone-refractory prostate cancer, Clin. Genitourin. Cancer 6 (2008)
Yu, L. Cao, S.M. Steinberg, J.B. Aragon-Ching, J. Venitz, E. Jones, C.C. Chen, W.D. 97–102.
Figg, A phase II clinical trial of sorafenib in androgen-independent prostate can- [121] L. Wu, D.C. Birle, I.F. Tannock, Effects of the mammalian target of rapamycin in-
cer, Clin. Cancer Res. 14 (2008) 209–214. hibitor CCI-779 used alone or with chemotherapy on human prostate cancer
[98] K.N. Chi, S.L. Ellard, S.J. Hotte, P. Czaykowski, M. Moore, J.D. Ruether, A.J. Schell, S. cells and xenografts, Cancer Res. 65 (2005) 2825–2831.
Taylor, C. Hansen, I. Gauthier, W. Walsh, L. Seymour, A phase II study of sorafe- [122] L.R. Molife, G. Attard, P.C. Fong, V. Karavasilis, A.H. Reid, S. Patterson, C.E. Riggs
nib in patients with chemo-naive castration-resistant prostate cancer, Ann. Jr., C. Higano, W.M. Stadler, W. McCulloch, D. Dearnaley, C. Parker, J.S. de Bono,
Oncol. 19 (2008) 746–751. Phase II, two-stage, single-arm trial of the histone deacetylase inhibitor
[99] J.B. Aragon-Ching, L. Jain, J.L. Gulley, P.M. Arlen, J.J. Wright, S.M. Steinberg, D. (HDACi) romidepsin in metastatic castration-resistant prostate cancer (CRPC),
Draper, J. Venitz, E. Jones, C.C. Chen, W.D. Figg, W.L. Dahut, Final analysis of a Ann. Oncol. 21 (2010) 109–113.
phase II trial using sorafenib for metastatic castration-resistant prostate cancer, [123] R. Ferrini, S.H. Woolf, American College of Preventive Medicine practice policy.
BJU Int. 103 (2009) 1636–1640. Screening for prostate cancer in American men, Am. J. Prev. Med. 15 (1998)
[100] R. Roskoski Jr., Sunitinib: a VEGF and PDGF receptor protein kinase and angio- 81–84.
genesis inhibitor, Biochem. Biophys. Res. Commun. 356 (2007) 323–328. [124] N. Bittner, G.S. Merrick, H. Andreini, W. Taubenslag, Z.A. Allen, W.M. Butler, R.L.
[101] O. Guerin, P. Formento, C. Lo Nigro, P. Hofman, J.L. Fischel, M.C. Etienne- Anderson, E. Adamovich, K.E. Wallner, Prebiopsy PSA velocity not reliable pre-
Grimaldi, M. Merlano, J.M. Ferrero, G. Milano, Supra-additive antitumor effect of dictor of prostate cancer diagnosis. Gleason score, tumor location, or cancer vol-
sunitinib malate (SU11248, Sutent) combined with docetaxel. A new therapeutic ume after TTMB, Urology 74 (2009) 171–176.
perspective in hormone refractory prostate cancer, J. Cancer Res. Clin. Oncol. 134 [125] S. Skvortsov, G. Schafer, T. Stasyk, C. Fuchsberger, G.K. Bonn, G. Bartsch, H.
(2008) 51–57. Klocker, L.A. Huber, Proteomics profiling of microdissected low- and high-grade
[102] A. Cumashi, N. Tinari, C. Rossi, R. Lattanzio, C. Natoli, M. Piantelli, S. Iacobelli, prostate tumors identifies Lamin A as a discriminatory biomarker, J. Proteome
Sunitinib malate (SU-11248) alone or in combination with low-dose docetaxel Res. 10 (2011) 259–268.
inhibits the growth of DU-145 prostate cancer xenografts, Cancer Lett. 270 [126] J.B. de Kok, G.W. Verhaegh, R.W. Roelofs, D. Hessels, L.A. Kiemeney, T.W. Aalders,
(2008) 229–233. D.W. Swinkels, J.A. Schalken, DD3(PCA3), a very sensitive and specific marker to
[103] M. Dror Michaelson, M.M. Regan, W.K. Oh, D.S. Kaufman, K. Olivier, S.Z. Michaelson, detect prostate tumors, Cancer Res. 62 (2002) 2695–2698.
B. Spicer, C. Gurski, P.W. Kantoff, M.R. Smith, Phase II study of sunitinib in men with [127] O.B. Goodman Jr., J.T. Symanowski, A. Loudyi, L.M. Fink, D.C. Ward, N.J. Vogelzang,
advanced prostate cancer, Ann. Oncol. 20 (2009) 913–920. Circulating tumor cells as a predictive biomarker in patients with hormone-
[104] G. Sonpavde, P.O. Periman, D. Bernold, D. Weckstein, M.T. Fleming, M.D. Galsky, sensitive prostate cancer, Clin. Genitourin. Cancer 9 (2011) 31–38.
W.R. Berry, F. Zhan, K.A. Boehm, L. Asmar, T.E. Hutson, Sunitinib malate for met- [128] S. Detchokul, A.G. Frauman, Recent developments in prostate cancer biomarker
astatic castration-resistant prostate cancer following docetaxel-based chemo- research: therapeutic implications, Br. J. Clin. Pharmacol. 71 (2011) 157–174.
therapy, Ann. Oncol. 21 (2010) 319–324. [129] C.O. Madu, Y. Lu, Novel diagnostic biomarkers for prostate cancer, J. Cancer 1
[105] D. Castellano, J.L. Gonzalez-Larriba, L.M. Anton-Aparicio, J. Cassinello, E. Grande, (2010) 150–177.
E. Esteban, J. Sepulveda, Experience in the use of sunitinib given as a single agent [130] G. Zhang, H. Zhang, Q. Wang, P. Lal, A.M. Carroll, M. de la Llera-Moya, X. Xu, M.I.
in metastatic chemoresistant and castration-resistant prostate cancer patients, Greene, Suppression of human prostate tumor growth by a unique prostate-
Expert. Opin. Pharmacother. 12 (2011) 2433–2439. specific monoclonal antibody F77 targeting a glycolipid marker, Proc. Natl.
[106] Y. Ou, M.D. Michaelson, L. Sengeløv, et al., Randomized, placebo-controlled, Acad. Sci. U. S. A. 107 (2010) 732–737.
phase III trial of sunitinib in combination with prednisone (SU+P) versus pred- [131] S.A. Tomlins, D.R. Rhodes, J. Yu, S. Varambally, R. Mehra, S. Perner, F. Demichelis,
nisone (P) alone in men with progressive metastatic castration-resistant pros- B.E. Helgeson, B. Laxman, D.S. Morris, Q. Cao, X. Cao, O. Andren, K. Fall, L.
tate cancer (mCRPC), ASCO Annual Meeting, 2011, Abstract 4515. Johnson, J.T. Wei, R.B. Shah, H. Al-Ahmadie, J.A. Eastham, S.E. Eggener, S.W.
[107] A.K. Gregorakis, E.H. Holmes, G.P. Murphy, Prostate-specific membrane antigen: Fine, K. Hotakainen, U.H. Stenman, A. Tsodikov, W.L. Gerald, H. Lilja, V.E. Reuter,
current and future utility, Semin. Urol. Oncol. 16 (1998) 2–12. P.W. Kantoff, P.T. Scardino, M.A. Rubin, A.S. Bjartell, A.M. Chinnaiyan, The role of
[108] P.M. Smith-Jones, S. Vallabhajosula, V. Navarro, D. Bastidas, S.J. Goldsmith, N.H. SPINK1 in ETS rearrangement-negative prostate cancers, Cancer Cell 13 (2008)
Bander, Radiolabeled monoclonal antibodies specific to the extracellular domain 519–528.
of prostate-specific membrane antigen: preclinical studies in nude mice bearing [132] N. Ozaki, M. Ohmuraya, M. Hirota, S. Ida, J. Wang, H. Takamori, S. Higashiyama,
LNCaP human prostate tumor, J. Nucl. Med. 44 (2003) 610–617. H. Baba, K. Yamamura, Serine protease inhibitor Kazal type 1 promotes prolifer-
[109] M.D. Henry, S. Wen, M.D. Silva, S. Chandra, M. Milton, P.J. Worland, A prostate- ation of pancreatic cancer cells through the epidermal growth factor receptor,
specific membrane antigen-targeted monoclonal antibody-chemotherapeutic Mol. Cancer Res. 7 (2009) 1572–1581.
conjugate designed for the treatment of prostate cancer, Cancer Res. 64 [133] F. Li, X. Ling, Survivin study: an update of “what is the next wave”? J. Cell. Physiol.
(2004) 7995–8001. 208 (2006) 476–486.
[110] M.D. Galsky, M. Eisenberger, S. Moore-Cooper, W.K. Kelly, S.F. Slovin, A. [134] N.K. Sah, Z. Khan, G.J. Khan, P.S. Bisen, Structural, functional and therapeutic
DeLaCruz, Y. Lee, I.J. Webb, H.I. Scher, Phase I trial of the prostate-specific biology of survivin, Cancer Lett. 244 (2006) 164–171.
membrane antigen-directed immunoconjugate MLN2704 in patients with pro- [135] G. Ambrosini, C. Adida, D.C. Altieri, A novel anti-apoptosis gene, survivin,
gressive metastatic castration-resistant prostate cancer, J. Clin. Oncol. 26 (2008) expressed in cancer and lymphoma, Nat Med 3 (1997) 917–921.
2147–2154. [136] M. Zhang, D.E. Latham, M.A. Delaney, A. Chakravarti, Survivin mediates resis-
[111] N.H. Bander, E.J. Trabulsi, L. Kostakoglu, D. Yao, S. Vallabhajosula, P. Smith-Jones, tance to antiandrogen therapy in prostate cancer, Oncogene 24 (2005)
M.A. Joyce, M. Milowsky, D.M. Nanus, S.J. Goldsmith, Targeting metastatic pros- 2474–2482.
tate cancer with radiolabeled monoclonal antibody J591 to the extracellular do- [137] M. Zhang, A. Ho, E.H. Hammond, Y. Suzuki, R.S. Bermudez, R.J. Lee, M. Pilepich,
main of prostate specific membrane antigen, J. Urol. 170 (2003) 1717–1721. W.U. Shipley, H. Sandler, L.Y. Khor, A. Pollack, A. Chakravarti, Prognostic value
[112] K. Kuroda, H. Liu, S. Kim, M. Guo, V. Navarro, N.H. Bander, Saporin toxin- of survivin in locally advanced prostate cancer: study based on RTOG 8610,
conjugated monoclonal antibody targeting prostate-specific membrane antigen Int. J. Radiat. Oncol. Biol. Phys. 73 (2009) 1033–1042.
has potent anticancer activity, Prostate 70 (2010) 1286–1294. [138] H. Wang, M.P. Holloway, L. Ma, Z.A. Cooper, M. Riolo, A. Samkari, K.S. Elenitoba-
[113] L. Whitesell, S.L. Lindquist, HSP90 and the chaperoning of cancer, Nat. Rev. Cancer Johnson, Y.E. Chin, R.A. Altura, Acetylation directs survivin nuclear localization
5 (2005) 761–772. to repress STAT3 oncogenic activity, J. Biol. Chem. 285 (2010) 36129–36137.
[114] A. Kamal, L. Thao, J. Sensintaffar, L. Zhang, M.F. Boehm, L.C. Fritz, F.J. Burrows, A [139] T. Nakahara, M. Takeuchi, I. Kinoyama, T. Minematsu, K. Shirasuna, A. Matsuhisa,
high-affinity conformation of Hsp90 confers tumour selectivity on Hsp90 inhib- A. Kita, F. Tominaga, K. Yamanaka, M. Kudoh, M. Sasamata, YM155, a novel
itors, Nature 425 (2003) 407–410. small-molecule survivin suppressant, induces regression of established human
[115] E.I. Heath, D.W. Hillman, U. Vaishampayan, S. Sheng, F. Sarkar, F. Harper, M. hormone-refractory prostate tumor xenografts, Cancer Res. 67 (2007)
Gaskins, H.C. Pitot, W. Tan, S.P. Ivy, R. Pili, M.A. Carducci, C. Erlichman, G. Liu, 8014–8021.
152 W. Fu et al. / Biochimica et Biophysica Acta 1825 (2012) 140–152
[140] G. Giaccone, P. Zatloukal, J. Roubec, K. Floor, J. Musil, M. Kuta, R.J. van [144] A. Berezov, Z. Cai, J.A. Freudenberg, H. Zhang, X. Cheng, T. Thompson, R. Murali,
Klaveren, S. Chaudhary, A. Gunther, S. Shamsili, Multicenter phase II trial M.I. Greene, Q. Wang, Disabling the mitotic spindle and tumor growth by target-
of YM155, a small-molecule suppressor of survivin, in patients with ad- ing a cavity-induced allosteric site of survivin, Oncogene (2011) (Electronic
vanced, refractory, non-small-cell lung cancer, J. Clin. Oncol. 27 (2009) publication ahead of print), doi:10.1038/onc.2011.377.
4481–4486. [145] M. Fishman, A changing world for DCvax: a PSMA loaded autologous dendritic
[141] K.D. Lewis, W. Samlowski, J. Ward, J. Catlett, L. Cranmer, J. Kirkwood, D. Lawson, cell vaccine for prostate cancer, Expert. Opin. Biol. Ther. 9 (2009) 1565–1575.
E. Whitman, R. Gonzalez, A multi-center phase II evaluation of the small mole- [146] S.S. Chang, V.E. Reuter, W.D. Heston, N.H. Bander, L.S. Grauer, P.B. Gaudin, Five
cule survivin suppressor YM155 in patients with unresectable stage III or IV different anti-prostate-specific membrane antigen (PSMA) antibodies confirm
melanoma, Invest New Drugs 29 (2011) 161–166. PSMA expression in tumor-associated neovasculature, Cancer Res. 59 (1999)
[142] A.W. Tolcher, A. Mita, L.D. Lewis, C.R. Garrett, E. Till, A.I. Daud, A. Patnaik, K. 3192–3198.
Papadopoulos, C. Takimoto, P. Bartels, A. Keating, S. Antonia, Phase I and phar- [147] M.J. Morris, C.R. Divgi, N. Pandit-Taskar, M. Batraki, N. Warren, A. Nacca, P.
macokinetic study of YM155, a small-molecule inhibitor of survivin, J. Clin. Smith-Jones, L. Schwartz, W.K. Kelly, S. Slovin, D. Solit, J. Halpern, A. Delacruz,
Oncol. 26 (2008) 5198–5203. T. Curley, R. Finn, A. O'Donoghue J, P. Livingston, S. Larson, H.I. Scher, Pilot trial
[143] M. Sun, W. Lou, J.Y. Chun, D.S. Cho, N. Nadiminty, C.P. Evans, J. Chen, J. Yue, Q. of unlabeled and indium-111-labeled anti-prostate-specific membrane antigen
Zhou, A.C. Gao, Sanguinarine suppresses prostate tumor growth and inhibits antibody J591 for castrate metastatic prostate cancer, Clin. Cancer Res. 11
survivin expression, Genes Cancer 1 (2010) 283–292. (2005) 7454–7461.