Abstract
Chemotherapy remains the major treatment option for castration-resistant prostate cancer (CRPC) and limited cytotoxic options are available. Inherent chemotherapy resistance occurs in half of all patients and inevitably develops even in those who initially respond. Docetaxel has been the mainstay of therapy for 6 years, providing a small survival benefit at the cost of significant toxicity. Cabazitaxel is a promising second-line agent; however, it is no less toxic, whereas mitoxantrone provides only symptomatic benefit. Multiple cellular pathways involving apoptosis, inflammation, angiogenesis, signalling intermediaries, drug efflux pumps and tubulin are implicated in the development of chemoresistance. A thorough understanding of these pathways is needed to identify biomarkers that predict chemotherapy resistance with the aim to avoid unwarranted toxicities in patients who will not benefit from treatment. Until recently, the search for predictive biomarkers has been disappointing; however, the recent discovery of macrophage inhibitory cytokine 1 as a marker of chemoresistance may herald a new era of biomarker discovery in CRPC. Understanding the interface between this complex array of chemoresistance pathways rather than their study in isolation will be required to effectively predict response and target the late stages of advanced disease. The pre-clinical evidence for these resistance pathways and their progress through clinical trials as therapeutic targets is reviewed in this study.
Introduction
Prostate cancer causes substantial morbidity and mortality worldwide (Ferlay et al. 2004) and is the second leading cause of cancer death in men in developed countries (AIHW & AACR 2008, Jemal et al. 2009). Metastatic prostate cancer initially responds to anti-androgen therapy; however, it eventually becomes resistant to hormonal manipulation. Chemotherapy remains the only treatment option in the setting of castration-resistant prostate cancer (CRPC) providing modest survival and palliative benefits. Only half of all patients will respond to docetaxel, a mitotic spindle poison that is the current mainstay of chemotherapy. Docetaxel improves median survival by 2 months at the cost of significant toxicity, particularly in this elderly patient population (Petrylak et al. 2004, Tannock et al. 2004). Mitoxantrone, a DNA intercalator, is less toxic but delivers only palliative benefits (Tannock et al. 1996, Kantoff et al. 1999). Inevitably, resistance to first-line chemotherapy will develop and the disease then becomes difficult to control. Although newer chemotherapeutics such as satraplatin and cabazitaxel have demonstrated activity as second-line agents, survival benefits remain modest with median overall survival just beyond 1 year (Sternberg et al. 2009b, De Bono et al. 2010c).
A thorough understanding of chemoresistance pathways and how they interact would facilitate two important outcomes. Identifying patients who will not benefit from chemotherapy prior to their exposure will avoid unnecessary toxicity and allow them to move on to alternative treatment options. Targets for further drug development may also arise. In this era of personalised cancer therapy, significant treatment advances have occurred through a better understanding of cytotoxic resistance and the heterogeneity among patients with the same disease. This concept has already proved successful across other cancer streams and is just the beginning of a new paradigm in cancer treatment. Indeed, many of the pathways implicated in prostate cancer chemoresistance may well be applicable to other cancer types.
There is an urgent need to identify markers and mechanisms of drug resistance to personalise treatment and improve survival. In this study, we have reviewed the current data regarding mechanisms of drug resistance in CRPC.
Apoptotic pathways
Clusterin
Clusterin is a glycoprotein whose mRNA is almost ubiquitously expressed in animal tissue. It occurs in two forms: an intracellular truncated nuclear form and a secreted heterodimeric disulphide linked form (sCLU). The nuclear form promotes apoptosis by translocating from the cytoplasm to the nucleus after cytotoxic events (Leskov et al. 2003), whereas the secreted form acts extracellularly to inhibit apoptosis by binding toxic molecules and targeting them for endocytic degradation (Wilson & Easterbrook-Smith 2000). In response to heat shock, sCLU is transcriptionally activated by heat-shock factor-1 and acts to chaperone and stabilise protein conformations at times of cell stress, similar to the small heat-shock proteins (HSPs; Wilson & Easterbrook-Smith 2000). sCLU also binds a variety of other molecules including Bax, which is normally activated by chemotherapeutic drugs leading to caspase activation and apoptosis, thereby preventing cell death (Zhang et al. 2005a). In prostate cancer cell lines, overexpression of clusterin increases resistance to apoptosis following treatment with chemotherapeutic agents such as taxanes and camptothecin. Furthermore, cytotoxic treatment of cells induces expression of clusterin, suggesting that this is a cytoprotective mechanism (Miyake et al. 2000d, Mizutani et al. 2006, Patterson et al. 2006). Clusterin expression may be regulated by the transcription factor Stat1. In hormone-resistant DU145 cells, Stat1 expression increases with docetaxel treatment, mirroring clusterin expression in these cells. In the docetaxel-resistant line, DU145-DR, inhibition of clusterin expression enhances docetaxel sensitivity (Patterson et al. 2006). In vivo, LNCaP xenografts that overexpress clusterin are less responsive to paclitaxel than controls (Miyake et al. 2000d). Clinically, sCLU expression is increased in radical prostatectomy specimens from patients who received neoadjuvant androgen ablation and docetaxel compared with those who had no preoperative therapy (P<0.001; Sowery et al. 2008).
Therapeutic anti-sense oligonucleotides (ASO) against clusterin have now been developed. Treatment of prostate cancer cell lines with clusterin ASO alone has no effect; however, it dramatically enhances the cytotoxic effect of chemotherapeutic agents, such as mitoxantrone and taxanes, when used in combination. A reduction in the IC50 of these agents by over 50% is seen even in previously chemoresistant cell lines (Miyake et al. 2000b,d,e, Sowery et al. 2008). These results have been replicated in prostate cancer xenografts with greater reductions in tumour volume and circulating PSA levels when chemotherapy was combined with clusterin ASO compared with chemotherapy alone (Miyake et al. 2000b,d,e, Springate et al. 2005, Sowery et al. 2008). In phase I trials, a clusterin ASO (OGX011) was well tolerated both as a single agent and in combination with weekly docetaxel (Chi et al. 2005a,b; Table 1). In a randomised phase II trial of 82 chemonaïve patients with CRPC where patients received docetaxel and OGX011/placebo, there was little difference in the primary endpoint, PSA response rate. However, there was a strong trend towards a substantial overall survival difference (27.5 vs 16.9 months; P=0.07; Chi et al. 2009b). In docetaxel-resistant patients, a phase II study where all patients received OGX011 and were randomised to receive docetaxel or mitoxantrone better PSA response rates were seen in the docetaxel arm (40 vs 27%) and overall survival mirrored this (14.7 vs 11.4 months; Saad et al. 2008). Phase II data of OGX011 in combination with docetaxel appears promising in both chemonaïve and chemoresistant CRPC patients. Phase III studies are now in progress to verify these results.
Reported clinical studies targeting apoptotic pathways in CRPC
Pathway | Inhibitors | Phase | Population | Intervention | Outcome | References |
---|---|---|---|---|---|---|
Clusterin | OGX011 | I | Localised PC | OGX011 | Tolerated | Chi et al. (2005a) |
I | Solid malignancies incl CRPC | OGX011+DTX | Tolerated | Chi et al. (2005b) | ||
II | Chemonaïve CRPC | OGX011±DTX (rand) | RR 58% | Chi et al. (2009b) | ||
II | DTX-resistant CRPC | OGX011+DTX | RR 40% | Saad et al. (2008) | ||
HSP | OGX427 | I | Solid malignancies incl CRPC | 17-AAG+Pac | Tolerated | Ramalingam et al. (2008) |
17-AAG | I | Solid malignancies incl CRPC | 17-AAG+DTX | Tolerated | Solit et al. (2005) | |
IPI504 | II | CRPC | IPI504 | No activity | Oh et al. (2009) | |
Bcl2 | Ob | I | CRPC | Ob+MTX | Tolerated | Chi et al. (2001) |
AT101 | I | Solid malignancies | Ob+DTX | Tolerated | Marshall et al. (2004) | |
I | Solid malignancies incl CRPC | Ob+Pac | Tolerated | Morris et al. (2005) | ||
I | Solid malignancies incl CRPC | Ob | Tolerated | Morris et al. (2002) | ||
I | CRPC | Ob+DTX | Tolerated | Tolcher et al. (2004) | ||
II | Chemonaïve CRPC | DTX±Ob (rand) | No activity | Sternberg et al. (2009a) | ||
I/II | Chemonaïve CRPC | AT101+DTX | RR 67% | MacVicar et al. (2009) | ||
I/II | DTX refractory CRPC | AT101+DTX | RR 18% | Poiesz et al. (2009) | ||
IAP | AEG35156 | I | Solid malignancies incl CRPC | AEG35156 | Tolerated | Dean et al. (2009) |
LY2181308 | I | Solid malignancies | LY2181308 | Tolerated | Talbot et al. (2008) | |
YM155 | I | Solid malignancies | YM155 | Tolerated | Satoh et al. (2009) | |
I | Solid malignancies incl CRPC | YM155 | Tolerated | Tolcher et al. (2008) |
PC, prostate cancer; DTX, docetaxel; CRPC, castration-resistant prostate cancer; rand, randomised; RR, response rate; HSP, heat-shock proteins; Ob, oblimersen; MTX, mitoxantrone; Pac, paclitaxel; IAP, inhibitors of apoptosis proteins.
Heat-shock proteins
HSPs are intracellular molecular chaperones that stabilise damaged proteins following a stressful insult such as heat-shock, hypoxia or cytotoxic therapy, thus inhibiting cell death. HSPs are classified according to their size in kilodaltons (e.g. HSP70 is 70 kDa in size). HSP27 prevents protein precipitation and sequesters cytochrome c and caspase 3, thus preventing activation of the caspase cascade and apoptosis. High levels, found in breast, prostate and ovarian cancers, are further induced by both hormone treatment and chemotherapy (Rocchi et al. 2004, Gleave et al. 2005). In PC3 cells and xenografts, blocking HSP27 expression by ASO and siRNA significantly enhances paclitaxel-induced apoptosis (Rocchi et al. 2004). A second-generation ASO OGX427 targeting HSP27 has entered phase I clinical trials.
HSP90 chaperones multiple proteins, including many signalling molecules that are important in oncogenesis (e.g. Akt, Raf1, Her2). This interaction prevents protein degradation by the ubiquitin proteasome pathway. HSP90 is overexpressed in prostate cancer tissue compared with normal prostate epithelium and may have a role in the evolution of prostate cancer to hormone resistance (Banerji 2009). 17-AAG, an analogue of geldanamycin, specifically inhibits HSP90 by blocking its ATP binding site. In hormone-resistant prostate cancer cells, 17-AAG prevents the ligand-independent nuclear translocation and activation of androgen receptors suggesting that HSP90 has a role in the development of androgen-independent disease (Saporita et al. 2007). In phase I clinical trials, 17-AAG was well tolerated in combination with docetaxel and paclitaxel (Solit et al. 2005, Musquire et al. 2007, Ramalingam et al. 2008; Table 1). A phase II study with 19 CRPC patients revealed that IPI504 (a water soluble form of 17-AAG) has no single-agent activity (Oh et al. 2009).
HSP70 and 72 have also been implicated in resistance to chemotherapy (Gabai et al. 2005, Ren et al. 2008) in prostate cancer cell lines; however, no therapeutic compounds directed against them are currently in trial. Therapy directed against HSPs has not been successful in early-phase trials; however, the full potential of these targets has not yet been thoroughly explored.
B-cell leukaemia/lymphoma 2
The protein products of the B-cell leukaemia/lymphoma 2 (Bcl2) gene family regulate apoptosis. Some members are pro-apoptotic (Bax, Bcl-XS, Bad), whereas others inhibit cell death (Bcl2, Bcl-xL, Mcl1). Bcl2, the prototypical member, exerts its anti-apoptotic effects by heterodimerising with pro-apoptotic members of the Bcl2 family (e.g. Bax) leading to their inhibition. This prevents apoptosis by inhibiting mitochondrial cytochrome c release and subsequent activation of the caspase cascade. Bcl2 up-regulation is implicated in the progression to androgen independence in prostate cancer (McDonnell et al. 1992, Raffo et al. 1995). Following treatment with paclitaxel, Bcl2 is phosphorylated in PC3 and LNCaP cells, inhibiting its anti-apoptotic action by preventing heterodimerisation with other members of the Bcl2 family and enhancing cell death (Haldar et al. 1996, Miyake et al. 2000a). Interestingly, DU145 cells, which do not express Bcl2, are inherently resistant to paclitaxel (Haldar et al. 1996). This suggests that the apoptotic action of taxane chemotherapy may rely, in part, on an interaction with Bcl2 rather than being mediated solely by microtubule stabilisation. Yoshino et al. (2006) further supported this theory with the finding that higher Bcl2 expression in prostate cancer tissue at baseline was an independent predictor for survival following taxane chemotherapy (P<0.01). Bcl-xL, another anti-apoptotic member, increases after androgen blockade, remains high in androgen-independent prostate cancer and correlates with increasing grade and stage. Forced overexpression of Bcl-xL in vitro leads to chemotherapy resistance while down-regulation enhances chemosensitivity (Lebedeva et al. 2000). High Mcl1 expression is seen in prostate cancer cell lines, high Gleason grade disease and bone metastases (Krajewska et al. 1996, Zhang et al. 2010). In vitro, Mcl1 overexpression has been implicated in resistance to cytokine-induced apoptosis and may also be involved in the anti-apoptotic action of interleukin 6 (IL6; Cavarretta et al. 2007, Dash et al. 2010).
Therapeutic ASOs directed against bcl2 family proteins have been developed and tested in the clinic. In pre-clinical studies, Bcl2 ASO is synergistic with taxane chemotherapy in both cell lines and xenografts (Miyake et al. 2000c, Leonetti et al. 2007). The combination of two ASOs directed against Bcl2 and Bcl-xL with paclitaxel appeared to be especially effective (Miyake et al. 2000c). Bispecific ASOs directed against both Bcl2 and Bcl-xL induce caspase activation and apoptosis in prostate cancer cell lines when used alone. Sensitivity to taxanes and mitoxantrone in vitro is also enhanced by these bispecific agents (Yamanaka et al. 2005, 2006). Oblimersen (G3139) is an ASO directed against the initiating sequence of Bcl2. Phase I trials of oblimersen demonstrated minimal toxicities both as a single agent and in combination with docetaxel or mitoxantrone (Chi et al. 2001, Morris et al. 2002, 2005, Marshall et al. 2004, Tolcher et al. 2004; Table 1). Disappointing results were seen in an EORTC first-line randomised phase II trial of 115 patients with CRPC given docetaxel with oblimersen or placebo. The end points were not met with a PSA response rate of 37% in the combination arm compared with 46% in the standard treatment arm (Sternberg et al. 2009a).
Multitargeted inhibitors of several Bcl family members are in clinical trials. A phase II study of docetaxel, prednisone and AT101, a small molecule inhibitor of Bcl2, Bcl-xL, Bcl-w and Mcl1, in 36 chemonaïve patients with CRPC demonstrated a PSA response rate of 67% (MacVicar et al. 2009). The same regimen used in a phase II trial with 34 patients with docetaxel refractory CRPC resulted in a PSA response rate of 18% with an objective response rate of 24% (Poiesz et al. 2009). A randomised phase II trial of docetaxel with AT101 or placebo in chemonaïve patients with metastatic CRPC is in process (clinicaltrials.gov ID: NCT00571675). There have been mixed results in early-phase clinical studies of Bcl inhibitors. Multitargeted agents have promising activity that needs to be verified in a randomised setting.
P53/murine double minute 2 protein
P53 is an extensively studied tumour suppressor protein that is activated during cell stress, including chemotherapy treatment or following DNA damage. Activated p53 regulates the transcription of genes to cause cell cycle arrest, DNA repair and occasionally apoptosis (Lane 1992). Murine double minute 2 protein (MDM2) acts as an oncogenic protein by targeting p53 for proteasomal degradation (Haupt et al. 1997, Honda et al. 1997), whereas MDM2 levels are increased by p53 in an autoregulatory loop (Bond et al. 2005). In vitro studies in prostate cancer cell lines suggest that MDM2 also acts via p53-independent pathways by modulating various cellular proteins including p21, Bax, pRb and Bcl2 (Zhang et al. 2003). MDM2 is amplified in many human cancers including prostate cancer and is a marker of advanced disease (Momand et al. 1998). In PC3, DU145 and LNCaP cells, ASO directed against MDM2 (AS-MDM2) cause dose-dependent apoptosis when used alone and increase chemosensitivity when used with paclitaxel and camptothecin. In PC3 and DU145 xenografts, AS-MDM2 exhibits anti-tumour activity alone and enhances sensitivity to paclitaxel and irinotecan chemotherapy (Wang et al. 2003, Zhang et al. 2003). Therapy directed against MDM2 has not yet entered clinical trials.
Inhibitors of apoptosis proteins
Inhibitors of apoptosis proteins (IAPs) are a family of five proteins (XIAP, survivin, HIAP1 and 2 and neuronal apoptosis inhibitory protein) that promote cell survival (Duckett et al. 1996, Liston et al. 1996, Chiou et al. 2003). They inhibit the caspase cascade and apoptosis by directly binding to caspases and cytochrome c. This occurs in response to a wide variety of apoptotic signals including chemotherapy treatment. IAPs also have a role in regulating cell cycle progression and modulating receptor-mediated signal transduction (Deveraux et al. 1998). They are induced by the transcription factor nuclear factor-kappa B (NF-κB) and via a feedback loop the HIAPs up-regulate NF-κB activity (LaCasse et al. 1998).
XIAP and survivin are the most well-understood IAPs with regard to carcinogenesis and chemoresistance. In vitro, overexpression of XIAP leads to paclitaxel resistance by preventing cleavage of pro-caspase 3 to caspase 3 and ultimately apoptosis (Nomura et al. 2003). An inhibitor of XIAP improves response to cisplatin by increasing caspase 3 activity in prostate cancer cell lines normally resistant to platinum therapy (DU145; Amantana et al. 2004). Survivin inhibition across several prostate cancer cell lines (LNCaP, DU145, PC3, C42B) increases sensitivity to docetaxel and etoposide (Hayashi et al. 2005, Rahman et al. 2009), whereas survivin overexpression increases paclitaxel resistance both in vitro and in vivo (Zhang et al. 2005b). In DU145 and PC3 xenografts, survivin inhibition either via an adenoviral anti-sense DNA vector or via a small molecule inhibitor leads to significant tumour regression alone and enhances the response to docetaxel and etoposide (Hayashi et al. 2005, Nakahara et al. 2007).
XIAP and survivin inhibitors have entered early-phase clinical trials (Table 1). An anti-sense inhibitor of XIAP (AEG35156) was well tolerated in a phase I trial in patients with refractory malignancies (Dean et al. 2009). Two phase I trials of this compound in combination with docetaxel in advanced solid tumours are ongoing (clinicaltrials.gov ID; NCT00372736 and NCT00357747). A survivin ASO (LY2181308) was also well tolerated in 24 patients with refractory tumours and no grade 3 or 4 toxicities were seen (Talbot et al. 2008). A randomised phase II trial of first-line therapy with docetaxel and LY2181308 or placebo in CRPC is in progress (NCT00642018). YM155, a small molecule inhibitor of survivin, was well tolerated in two phase I trials (Tolcher et al. 2008, Satoh et al. 2009) and preliminary activity was suggested in patients with docetaxel refractory CRPC (Tolcher et al. 2008). A phase I/II study in advanced CRPC of docetaxel in combination with YM155 is ongoing (NCT00514267). Agents targeting IAPs have recently entered the early stages of clinical testing and evidence of therapeutic activity is not yet available.
Inflammation
Nuclear factor-kappa B
There is mounting evidence that the inflammatory response plays an integral role at all stages of prostate carcinogenesis from prostate intraepithelial neoplasia (PIN; De Marzo et al. 2007) to the development of hormonal therapy and chemotherapy resistance. NF-κB is a transcription factor central to the inflammatory response. In the cytoplasm, it is bound to inhibitory proteins (IκBs) that render it inactive; however, on stimulation of the IκB kinase (IKK) complex, these inhibitory proteins are proteolysed leaving NF-κB free to translocate to the nucleus and activate a wide spectrum of genes for interleukins growth factors, stress response elements (including cyclooxygenase-2) and anti-apoptotic proteins (Paule et al. 2007). Increased expression of NF-κB is seen in the progression from normal prostatic epithelium to high-grade PIN to invasive prostate cancer (Sweeney et al. 2004). NF-κB is activated by tumour necrosis factor α (TNF-α; Singh & Aggarwal 1995) promoting cell survival in androgen-independent cell lines. Conversely, in androgen-dependent LNCaP cells, TNF-α causes apoptosis with no NF-κB interaction (Zhao et al. 1992, Chopra et al. 2004, Srinivasan et al. 2010). Many cytotoxic agents including taxanes, cisplatin, 5-fluorouracil and doxorubicin also induce NF-κB (Chuang et al. 2002, Li et al. 2005, Shaikh et al. 2008). It is constitutively activated in androgen-independent prostate cancer cell lines, PC3 and DU145, and is associated with elevated IL6 production in conditioned media. In contrast, androgen-dependent LNCaP cells do not exhibit increased NF-κB activity or IL6 secretion. Indirect inhibition of NF-κB with an IKK complex inhibitor enhances docetaxel sensitivity in PC3 and DU145 cells, while having no effect on LNCaP cells (Domingo-Domenech et al. 2006). Inhibitors of NF-κB derived from natural compounds, such as curcumin, genistein and docosahexaenoic acid, display synergistic effects with cytotoxic agents (taxanes, cisplatin and 5-fluorouracil) in prostate cancer cell lines (Hour et al. 2002, Li et al. 2005, Shaikh et al. 2008). No specific NF-κB inhibitors have yet entered prostate cancer clinical trials.
Interleukin 6
IL6 is an important downstream cytokine implicated in many aspects of prostate cancer progression. Androgen-independent prostate cancer cell lines (PC3 and DU145) constitutively produce IL6, which acts as an autocrine growth factor and is at least in part responsible for the transition of cells from an androgen-dependent to androgen-independent state (Hobisch et al. 1998, Chung et al. 1999, Chen et al. 2000, Wallner et al. 2006). In vitro, inhibiting IL6 enhances chemosensitivity, whereas exogenous IL6 inhibits cytotoxic drug-induced apoptosis (Borsellino et al. 1995, Pu et al. 2004). These effects may be mediated via the Bcl and Stat signalling pathways (Pu et al. 2004).
In clinical cohorts of CRPC patients receiving docetaxel chemotherapy, elevated baseline of serum IL6 levels inversely correlate with response (P=0.039), time to progression (P=0.023) and prostate cancer-specific and overall survival (P<0.001; Domingo-Domenech et al. 2007, Visa et al. 2009). Interestingly, increased NF-κB staining in cancer tissue directly correlated with elevated serum IL6 levels in one cohort, suggesting that IL6 may serve as a surrogate measure for NF-κB activity (P=0.009; Domingo-Domenech et al. 2007). Elevated baseline of serum levels of C-reactive protein, an acute phase protein released by the liver in response to rising IL6 levels, also inversely correlate with PSA response and overall survival in a clinical cohort of 160 CRPC patients undergoing docetaxel therapy (P<0.0001; Beer et al. 2008).
A chimeric anti-IL6 antibody (CNTO 328) has been shown to inhibit prostate cancer xenograft growth (Smith & Keller 2001, Wallner et al. 2006) and has entered clinical trials (Table 2). A phase I trial of first-line docetaxel and CNTO 328 in 38 patients with CRPC was well tolerated (Hudes et al. 2009). In a phase II SWOG study, 54 patients with CRPC who had failed prior taxane therapy received CNTO 328 alone. The compound was again well tolerated with modest clinical activity (PSA response rate 3.7%, RECIST stable disease rate 21%; Pinski et al. 2009). A randomised phase II study of CNTO 328 or placebo in combination with mitoxantrone was terminated early due to more deaths in the experimental arm (De Bono et al. 2010a). Anti-IL6 therapy has yielded disappointing results and failed to progress beyond early-phase clinical trials.
Reported clinical studies targeting inflammatory pathways in CRPC
Pathway | Inhibitors | Phase | Population | Intervention | Outcome | References |
---|---|---|---|---|---|---|
IL6 | CNTO 328 | I | Chemonaïve CRPC | CNTO 328+DTX | Tolerated | Hudes et al. (2009) |
II | DTX-resistant CRPC | CNTO 328 | RR 4% | Pinski et al. (2009) | ||
II | DTX-resistant CRPC | MTX±CNTO 328 (rand) | Terminated early | De Bono et al. (2010a) |
IL6, interleukin 6; DTX, docetaxel; CRPC, castration-resistant prostate cancer; MTX, mitoxantrone; RR, response rate; rand, randomised.
Interleukin 8
IL8 is also implicated in prostate cancer progression. Secreted by leucocytes and tumour cells and acting via two high-affinity membrane receptors, CXCR1 and 2, IL8 enhances angiogenesis by increasing expression of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF; Xie 2001). IL8 expression is primarily regulated by NF-κB and activator protein-1 (Brat et al. 2005). Although IL8 is only secreted by androgen-independent prostate cancer cell lines, its receptors are expressed similarly by all prostate cancer cells. Following IL8 overexpression, androgen-dependent cell lines (LNCaP and LAPC4) were rendered less sensitive to androgen blockade and docetaxel. The resistance to docetaxel was reversed by concomitant treatment with NF-κB and src inhibitors suggesting that IL8 activation of these pathways is involved in chemoresistance (Araki et al. 2007). PC3 and DU145 cells also exhibit increased NF-κB activity when exposed to exogenous IL8. NF-κB activity is up-regulated by treatment with oxaliplatin leading to increased transcription of IL8 and CXCR2 genes. Pre-treatment with a CXCR2 antagonist (AZ10397767) reduces oxaliplatin-induced NF-κB activation and enhances chemosensitivity (Wilson et al. 2008). IL8 may also be implicated in the relative chemoresistance of hypoxic cancer cells. When PC3 cells are rendered hypoxic, expression of IL8 and CXCR 1 and 2 is enhanced. Following treatment with etoposide, hypoxic PC3 cells are less sensitive than normoxic cells; however, siRNA inhibition of IL8 normalises the response of hypoxic cells to chemotherapy suggesting that IL8 plays a central role in this differential response (Maxwell et al. 2007).
Macrophage inhibitory cytokine 1
Macrophage inhibitory cytokine 1 (MIC1), also known as growth differentiation factor 15, is a member of the transforming growth factor β superfamily. MIC1 appears to have anti-tumour activity as it plays a role in cell cycle arrest, induction of apoptosis and anti-angiogenesis. High levels are only normally found in the placenta; however, expression increases in disease states such as acute injury, inflammation and malignancy. In particular, high levels are found in metastatic prostate, colon and breast cancers (Bauskin et al. 2006). In early low-grade prostate cancer, the unprocessed form of MIC1 found in stromal stores inversely correlates with the risk of future relapse (Bauskin et al. 2005), whereas serum MIC1 levels in combination with PSA improve on the specificity of PSA alone in screening for prostate cancer (Brown et al. 2006). In PC3 and DU145 cells, exposure to docetaxel and mitoxantrone leads to MIC1 overexpression (Huang et al. 2007). On proteomic analysis comparing docetaxel-sensitive (PC3) and docetaxel-resistant (PC3-Rx) cells, there was pronounced overexpression of MIC1 in PC3-Rx cells (Zhao et al. 2009). PC3 cells exposed to recombinant MIC1 exhibited increased docetaxel resistance, whereas MIC1 inhibition in PC3-Rx cells via siRNA restored docetaxel sensitivity (Zhao et al. 2009). In a cohort of 38 men with CRPC receiving docetaxel or mitoxantrone chemotherapy, an increase in circulating MIC1 after the first cycle of chemotherapy was inversely correlated with PSA response (P=0.006) and overall survival (P=0.002; Zhao et al. 2009). The identification of this promising biomarker may herald the beginning of our ability to personalise therapy and detect chemotherapy resistance earlier in the treatment course.
Vasculature
Dysregulated angiogenesis associated with malignancy may contribute to chemotherapy resistance. Abnormal vascular organisation, altered stromal composition and increased permeability of blood vessels result in increased interstitial fluid pressure and ultimately limited drug permeability (Heldin et al. 2004). Transient hypoxia and nutrient deprivation of cancer cells may also result in reduced cell proliferation and impaired chemotherapy sensitivity (Bellone et al. 2008). By normalising tumour vasculature, delivery of oxygen and cytotoxic drugs to tumour tissue may be improved (Jain 2005). There are a variety of drugs that act to modulate angiogenesis, including targeted agents and more conventional chemotherapeutics.
Agents targeted against VEGF include bevacizumab, sunitinib, sorafenib and aflibercept. The VEGF family comprises at least seven members (namely A, B, C, D, E, F and placental growth factor) with VEGFA playing the most important role in angiogenesis. It binds to two tyrosine kinase receptors (VEGFR1 and 2; Epstein 2007). In 197 patients on a phase III study in CRPC, high pre-treatment plasma VEGF levels correlated with reduced overall survival (George et al. 2001). In vitro, docetaxel has inherent anti-angiogenic properties that are negated when endothelial cells are stimulated by VEGF and bFGF. When a human breast adenocarcinoma xenograft (MCF7) is treated with docetaxel and a VEGF inhibitor, this protective effect is overcome, resulting in synergistic activity (Sweeney et al. 2001). The murine equivalent of bevacizumab, a humanised monoclonal antibody directed against VEGFA (Presta et al. 1997), inhibits tumour growth and metastases in CRPC xenografts when used as a single agent (Borgstrom et al. 1998, Melnyk et al. 1999) and is synergistic with paclitaxel (Fox et al. 2002).
Clinically, bevacizumab has no significant activity in prostate cancer as a single agent (Reese et al. 2001; Table 3); however, it appears promising in combination with cytotoxic agents. In 20 patients on a phase II trial with chemotherapy refractory CRPC, docetaxel and bevacizumab resulted in a PSA response rate of 55% (including four patients who did not respond initially to docetaxel alone; Di Lorenzo et al. 2008). Two first-line phase II trials revealed PSA response rates over 80% with bevacizumab and docetaxel combined with thalidomide or estramustine (Picus et al. 2003, Ning et al. 2008). Unfortunately, a large randomised phase III trial (CALGB 90401) of first-line docetaxel and bevacizumab or placebo in 1050 CRPC patients revealed no overall survival benefit and was associated with greater morbidity and mortality (Kelly et al. 2010).
Reported clinical studies targeting vascular pathways in CRPC
Pathway | Inhibitors | Phase | Population | Intervention | Outcome | References |
---|---|---|---|---|---|---|
VEGF | Bevacizumab | I | Chemonaïve CRPC | Bv+Ev+DTX | Tolerated | Gross et al. (2009) |
II | CRPC | Bv | No activity | Reese et al. (2001) | ||
II | DTX-resistant CRPC | Bv+DTX | RR 55% | Di Lorenzo et al. (2008) | ||
II | Chemonaïve CRPC | Bv+DTX+E | RR 65% | Picus et al. (2003) | ||
II | Chemonaïve CRPC | Bv+DTX+Th | RR 88% | Ning et al. (2008) | ||
III | Chemonaïve CRPC | DTX±Bv (rand) | No OS benefit | Kelly et al. (2010) | ||
Sunitinib | II | CRPC | Su | RR 6% | Dror Michaelson et al. (2009) | |
II | DTX-resistant CRPC | Su | RR 24% | Castellano et al. (2010) | ||
Sorafenib | I | DTX-resistant CRPC | So+MTX or DTX | Tolerated | Nabhan et al. (2010) | |
II | Chemonaïve CRPC | So+DTX | RR 46% | Cetnar et al. (2009) | ||
II | CRPC | So | Discordant response | Dahut et al. (2006) | ||
Aflibercept | I | Solid malignancies | Af+DTX | Tolerated | Isambert et al. (2008) | |
Thalidomide | II | CRPC | Th (LD versus HD) | RR 18% (LD) versus 0% (HD) | Figg et al. (2001b) | |
II | Chemonaïve CRPC | DTX±Th (rand) | RR 53 vs 35% | Figg et al. (2001a) | ||
Lenalidomide | I | Solid malignancies incl CRPC | Ln | Tolerated | Tohnya et al. (2006) | |
I | CRPC | Ln+DTX | Tolerated | Moss et al. (2007) |
MTX, mitoxantrone; DTX, docetaxel; Ev, everolimus; rand, randomised; CRPC, castration-resistant prostate cancer; RR, response rate; Bv, bevacizumab; E, estramustine; OS, overall survival; Su, sunitinib; So, sorafenib; Af, aflibercept; Th, thalidomide; LD, low dose; HD, high dose; Ln, lenalidomide.
Sunitinib is a small molecule receptor tyrosine kinase inhibitor that has many targets, including VEGFR and platelet-derived growth factor receptor (PDGFR). Synergistic activity was seen in combination with docetaxel in CRPC xenografts (Cumashi et al. 2008, Guerin et al. 2008), whereas in early clinical studies, single-agent sunitinib exhibited some activity (Dror Michaelson et al. 2009, Castellano et al. 2010). Phase I and II trials combining sunitinib and docetaxel in CRPC are ongoing (clinicaltrials.gov ID; NCT00137436, NCT00879619) whereas a phase II trial of maintenance sunitinib following response to docetaxel is in progress (NCT00550810). A randomised phase III placebo-controlled trial of sunitinib alone in docetaxel refractory CRPC is also in the recruitment phase (NCT00676650).
Sorafenib is another small molecule receptor tyrosine kinase inhibitor that targets VEGF, PDGFR and Raf. As a single agent, sorafenib inhibits growth of both androgen-sensitive (LNCaP) and androgen-resistant (PC3) cells in vitro. Further studies on chemotherapy-resistant cells are in progress (Culig et al. 2009). Clinically, a phase II study of first-line sorafenib and docetaxel in metastatic CRPC revealed a PSA response rate comparable to docetaxel alone (Cetnar et al. 2009); however, there is a suggestion that the PSA response rate does not correlate well with objective responses following sorafenib treatment (Dahut et al. 2006). More promising is an ongoing phase I/II study of the addition of sorafenib to docetaxel or mitoxantrone therapy at the time of disease progression. Preliminary results in 16 patients demonstrated a PSA decline in 43% of patients with a median survival of 8 months (Nabhan et al. 2010). This indicates that sorafenib may play some role in overcoming chemotherapy refractory disease.
Aflibercept (VEGF trap) is a soluble decoy receptor that binds to circulating VEGF via the human VEGFR extracellular domains fused to the Fc portion of human IgG1. This was well tolerated in combination with docetaxel in a phase I trial in chemotherapy refractory-advanced solid tumours (Isambert et al. 2008). A randomised phase III trial of first-line docetaxel and aflibercept or placebo in CRPC has completed enrolment (VENICE study, clinicaltrials.gov ID NCT00519285).
Biological therapies targeted at VEGF have demonstrated promising results in phase I and II clinical trials. Phase III data with first-line bevacizumab and docetaxel was disappointing; however, we await results of randomised trials with other anti-VEGF agents.
Thalidomide and its analogue lenalidomide are cytotoxic drugs with anti-angiogenic properties. While the anti-angiogenic mechanism is still uncertain, a reduction in VEGF and bFGF may play a role (Aragon-Ching & Dahut 2008). A phase II CRPC clinical trial of high- versus low-dose thalidomide revealed very poor tolerance of the high-dose and an 18% PSA response rate in the low-dose arm (Figg et al. 2001b). A randomised phase II study of first-line weekly docetaxel and thalidomide or placebo in 75 CRPC patients demonstrated favourable PSA response rates of 53% in the combined arm versus 37% with docetaxel alone. The overall survival was also improved with combined therapy (25.9 vs 14.7 months). Patients required prophylactic low molecular weight heparin due to an increased incidence of thromboembolism in the combination therapy arm (Figg et al. 2001a, 2005).
Compared to thalidomide, lenalidomide has a better safety profile and is a more potent angiogenic inhibitor (Tohnya et al. 2006). It was well tolerated both alone and in combination with docetaxel in phase I trials (Tohnya et al. 2006, Moss et al. 2007). Phase II trials in CRPC of lenalidomide in combination with taxanes and/or bevacizumab are currently recruiting (clinicaltrials.gov ID; NCT00933426, NCT00942578). A randomised phase III trial of first-line docetaxel and lenalidomide or placebo in CRPC is also in the recruitment phase (clinicaltrials.gov ID; NCT00988208).
Signalling intermediaries
Stat
The signal transducers and activator of transcription (Stat) proteins are a family of seven cytoplasmic transcription factors (Stat 1, 2, 3, 4, 5A, B and 6) that dimerise and translocate to the nucleus on activation. They regulate gene expression to influence differentiation, proliferation, apoptosis and angiogenesis. In stress-induced responses, they are activated via cytokine signalling to modulate pro- and anti-apoptotic genes (Stephanou & Latchman 2003). Stat1 was initially thought to act as a tumour suppressor as Stat1-deficient mice developed early aggressive tumours and Stat1-deficient cancer cells were more resistant to chemotherapy (Stephanou & Latchman 2003). However, in prostate cancer cell lines, Stat1 appears to be associated with docetaxel resistance. Stat1 is overexpressed in docetaxel-resistant DU145-DR and PC3-DR cell lines. Following docetaxel treatment of sensitive DU145 cells, Stat1 expression increases over time, mirroring the reduction in apoptosis. SiRNA targeting Stat1 rendered these cells more sensitive to docetaxel-induced apoptosis. As discussed earlier, these actions appeared to be linked to clusterin with clusterin expression decreasing following treatment with Stat1 siRNA in DU145-DR cells (Patterson et al. 2006). Stat3 also appears to be involved in docetaxel resistance in prostate cancer cell lines. This action may be due to induction of PIM1 kinase, a serine threonine kinase that promotes cell survival. Following treatment with docetaxel, PIM1 kinase is overexpressed in DU145 cells and xenografts leading to reduced sensitivity to docetaxel. It has been postulated that Stat3 phosphorylation, which is promoted by docetaxel treatment, leads to overexpression of PIM1 kinase, increased NF-κB activity and ultimately drug resistance (Zemskova et al. 2008). Interestingly, Stat3 is also activated by IL6 (Stephanou & Latchman 2003), linking this further with the inflammatory process. Stat inhibition has not yet progressed into clinical studies.
Insulin-like growth factors
The insulin-like growth factor (IGF) axis includes circulating peptide growth factors (IGF1 and 2), transmembrane receptors (IGF1R and IIR) and six IGF binding proteins (IGFBP1–6). It affects carbohydrate and protein metabolism and regulates cellular proliferation, apoptosis and differentiation (Chi et al. 2009a). Circulating IGF and IGFBP levels correlate with stage and grade of prostate cancer (Figueroa et al. 1998, Chan et al. 2002), while IGFBP overexpression is associated with progression to androgen independence (Miyake et al. 2000b,e). Humanised monoclonal antibodies directed against the IGF1 receptor have entered the clinical arena in an attempt to alter chemotherapy resistance. A randomised phase II study testing docetaxel and one such Ab, CP751,871/placebo, is currently recruiting, including chemotherapy naïve and docetaxel refractory arms (clinicaltrials.gov ID; NCT00313781). Two phase II trials of IMC-A12 are underway. A single-arm study will test IMC-A12 alone in chemotherapy naïve CRPC patients (NCT00520481) and a second study includes docetaxel-resistant CRPC patients randomised to mitoxantrone and IMC-A12 or IMC1121B, an anti-VEGFR2 fully human monoclonal antibody (NCT00683475). Early-phase testing of IGF receptor inhibitors has just commenced with no data available to date.
Phosphoinositide 3′-kinase/Akt/mammalian target of rapamycin pathway
Dysregulation of intracellular pathways involving the kinases phosphoinositide 3′-kinase (PI3K), Akt and mammalian target of rapamycin (mTOR) leads to enhanced cell survival, cell cycle progression, neoplastic transformation and chemotherapy resistance. In normal circumstances, the tumour suppressor protein, PTEN, negatively regulates this pathway; however, this function is lost in up to 80% of prostate cancers leading to constitutive activation of Akt. Akt prevents apoptosis by phosphorylating and inhibiting pro-apoptotic factors such as BAD, pro-caspase 9, FKHR and Bim. It also inhibits release of the caspase cascade inducer, cytochrome c. PI3K activation may promote the development of chemoresistance by up-regulating the expression of multidrug resistance protein 1 (MRP1), a drug efflux pump. Both PI3K and Akt indirectly activate mTOR causing overexpression of various proto-oncogenes and growth factors, including c-myc, cyclin D1 and VEGF (Lee et al. 2008, Meric-Bernstam & Gonzalez-Angulo 2009).
In vitro, PTEN expressing DU145 cells are more sensitive to doxorubicin and paclitaxel chemotherapy than PC3 cells, which do not express PTEN. However, the chemosensitivity of PC3 cells is restored when the pathway is inhibited, either by PTEN transfection or by direct mTOR inhibition with rapamycin (Grunwald et al. 2002, Lee et al. 2004). Long-term androgen ablated cells, LNCaP-abl, are resistant to chemotherapy, but following treatment with a PI3K inhibitor, LY294002, chemo-sensitivity is restored, further highlighting the potential of this pathway as a target for treatment (Pfeil et al. 2004). Everolimus, an mTOR inhibitor, has exhibited some clinical activity in combination with bevacizumab and docetaxel as first-line treatment in CRPC (Gross et al. 2009; Table 3). A phase II trial of first-line docetaxel and everolimus in CRPC is ongoing (clinicaltrials.gov ID; NCT00459186). Another mTOR inhibitor, ridaforolomus, has completed phase II testing in the docetaxel-resistant setting, with results pending (clinicaltrials.gov ID; NCT00110188). Activity of mTOR inhibitors in CRPC is yet to be established.
Platelet-derived growth factor
PDGF receptors are overexpressed in prostate cancers compared with benign prostatic epithelium making them a potential target for enhancing chemosensitivity. However, the level of overexpression may be modest with one study showing PDGFR expression in only 16% of advanced CRPC tumour tissue (Hofer et al. 2004). Imatinib is a PDGFR tyrosine kinase inhibitor that has been assessed in both pre-clinical and clinical studies in CRPC. When multidrug-resistant prostate cancer cells (PC–3MM2–MDR) were treated in vitro with imatinib and paclitaxel, resistance to both drugs was seen. However, in PC–3MM2–MDR mouse tibial xenografts, treatment with imatinib alone and in combination with paclitaxel led to reduced bone tumour incidence, reduced tumour weight, reduced bone lysis, less lymph node metastases and decreased mean vessel density. This suggested that the target for imatinib was the tumour-associated endothelial cells rather than the prostate tumour cells themselves (Kim et al. 2006). Cell growth of PC3, DU145 and LNCaP cells is inhibited with imatinib; however, antagonistic effects were seen when combined with docetaxel, particularly in PC3 cells (Kubler et al. 2004). Clinically, the combination of cytotoxic therapy with imatinib has not been promising (Table 4). Two single-arm phase II trials combining docetaxel and imatinib revealed only modest response rates and significant toxicity (Gillison et al. 2009, Gomez-Pinillos et al. 2009). A larger randomised, double-blind phase II trial with 104 CRPC patients with bone metastases and no prior taxane exposure tested docetaxel and imatinib/placebo. The primary end point, time to progression, was worse in the experimental arm (4.4 vs 5.3 months). Again, toxicity was significant, particularly gastrointestinal (Mathew et al. 2006). Interestingly, an associated biomarker study revealed that in the standard treatment arm, reduced PDGFR phosphorylation in peripheral blood mononuclear cells was associated with poorer outcomes including decreased progression-free and overall survival (P=0.04). It was suggested that this may be a marker or mechanism for docetaxel resistance; however, further studies have not yet been published (Mathew et al. 2008).
Reported clinical studies targeting other pathways in CRPC
Pathway | Agents | Phase | Population | Intervention | Outcome | References |
---|---|---|---|---|---|---|
Multidrug resistance | Laniquidar | I | Solid malignancies | La+DTX | Tolerated | Van Zuylen et al. (2002) |
Biricodar | I | Solid malignancies | Zo+DTX | Tolerated | Fracasso et al. (2004) | |
Zosuquidar | II | Chemonaïve CRPC | Bi+MTX | RR 30% | Rago et al. (2003) | |
Cabazitaxel | I | Solid malignancies | Ca | Tolerated | Gelmon et al. (2000) | |
I | Solid malignancies incl CRPC | Ca | Tolerated | Mita et al. (2009) | ||
III | DTX-resistant CRPC | Ca or MTX (rand) | OS 15.1 vs 12.7 months | De Bono et al. (2010b) | ||
Tubulin | Epothilones | I/II | DTX-resistant CRPC | Ix+MTX | RR 31% | Rosenberg et al. (2009) |
II | Chemonaïve CRPC | Ix | RR 32% | Wilding et al. (2008) | ||
II | DTX-resistant CRPC | Ix | RR 22% | Wilding et al. (2008) | ||
II | Chemonaïve CRPC | Ix | RR 33% | Hussain et al. (2005) | ||
II | CRPC | Pa | RR 13% | Hussain et al. (2009) | ||
II | DTX-resistant CRPC | Pa | RR 45% | Beardsley et al. (2009) | ||
PDGF | Imatinib | II | CRPC | Im+DTX | RR 41% | Gillison et al. (2009) |
II | CRPC | Im+DTX | RR 47% | Gomez-Pinillos et al. (2009) | ||
II | Chemonaïve CRPC | DTX±Im | No difference | Mathew et al. (2006) |
MTX, mitoxantrone; DTX, docetaxel; CRPC, castration-resistant prostate cancer; RR, response rate; OS, overall survival; La, laniquidar; Zo, zosuquidar; Bi, biricodar; Ca, cabazitaxel; Ix, ixabepilone; Pa, patupilone; PDGF, platelet derived growth factor; Im, imatinib.
Multidrug resistance proteins
MDRPs, including P-glycoprotein (P-gp; encoded by the MDR1 gene) and MRP1, are ATP binding cassette (ABC) transporters in cell membranes of the biliary tract, intestinal epithelium, the blood–brain barrier and tumours (Bradshaw & Arceci 1998). They act as drug efflux pumps with a wide range of substrates, including docetaxel and mitoxantrone (van Zuylen et al. 2000). In prostate cancer cell lines, P-gp appears to be variably related to chemotherapy resistance. Chemoresistant PC3 cells do not overexpress P-gp, whereas resistant DU145 cells exhibit both overexpression of the protein and restored chemo-sensitivity with P-gp inhibition (Makarovskiy et al. 2002, Takeda et al. 2007). Furthermore, exposure to mitoxantrone and docetaxel results in increased multidrug-resistant protein expression in chemo-sensitive prostate cancer cell lines (Sanchez et al. 2009). Although MDR1 is well understood, MRP1 is also emerging as an important element in prostate cancer chemoresistance (Zalcberg et al. 2000, Lee et al. 2008). Up-regulated MRP1 expression is found in chemoresistant DU0.03 and PC0.03 cells with no up-regulation in P-gp (Zalcberg et al. 2000). Both p53 and PIM1 kinase appear to be involved in modulating ABC transporter expression and chemoresistance, providing potential future targets for treatment (Sullivan et al. 2000, Xie et al. 2008). In a cohort of 73 CRPC patients receiving docetaxel±thalidomide, genotyping for the ABCB1 transporter was performed. Particular variants of this gene were significantly correlated with overall survival (P=0.0048) and toxicity (including neuropathy and neutropaenia) suggesting that specific ABC transporter genotypes may predict outcome with chemotherapy (Sissung et al. 2008).
Therapeutic manipulation of MDR proteins was initially attempted with co-administration of classical MDR modulators (such as verapamil, cyclosporine A and valspodar) and chemotherapeutic agents. Unfortunately, these drugs were disappointing due to their toxicity and unpredictable effects on pharmacokinetics. Since then, third-generation modulators that cause minimal pharmacokinetic interference have been developed. P-gp inhibitors, including laniquidar, biricodar and elacridar, are well tolerated in combination with chemotherapy for CRPC in phase I trials, however exhibited minimal clinical activity on phase II analysis (van Zuylen et al. 2002, Lokiec et al. 2003, Rago et al. 2003, Fracasso et al. 2004; Table 4).
Cabazitaxel is a novel taxane, which avoids cellular extrusion due to poor affinity for P-gp. In vitro, potent cytotoxic effects were achieved in a variety of cell lines, including those with docetaxel resistance due to P-gp overexpression. These results were mirrored in vivo, including in a CRPC xenograft model (DU145; Mita et al. 2009). In a recent phase III study, 755 men with docetaxel-resistant CRPC were randomised to receive cabazitaxel or mitoxantrone. Overall survival was improved in the experimental arm (15.1 vs 12.7 months; P<0.0001; De Bono et al. 2010b) making cabazitaxel a promising second-line chemotherapy option.
Tubulin
Growing pre-clinical evidence suggests that resistance to taxane chemotherapy in CRPC may be attributed to changes in β-tubulin isotypes, the primary target of these drugs. There are at least seven isotypes of β-tubulin with the predominant type in most normal tissues being isotype I (Luduena 1998). Paclitaxel resistance is associated with a switch from class I to class III β-tubulin in multiple cancer cell lines (Kamath et al. 2005). In CRPC cell lines, an increase in isotypes III and IV correlates with docetaxel and paclitaxel resistance (Ranganathan et al. 1998, Makarovskiy et al. 2002). Oestrogen treatment of hormone-dependent LNCaP cells suppressed expression of β tubulin IVa and led to improved docetaxel sensitivity in xenografts (Montgomery et al. 2005). In a phase II clinical trial with 29 CRPC patients, first-line diethylstilboestrol and docetaxel led to a 75% PSA response rate (Montgomery et al. 2006).
Novel agents targeting tubulin have recently been developed. Epothilones are cytotoxic macrolides, which, like taxanes, prevent tubulin depolymerisation causing mitotic arrest and apoptosis. While epothilones bind tubulin at the same site as taxanes, they have a more potent effect on polymerisation. This results in greater inhibition of tumour growth in prostate cancer xenografts than paclitaxel (Newman et al. 2001). Epothilones are effective in some taxane-resistant settings. In tumours overexpressing P-gp and in many with taxane-resistant tubulin mutations, epothilones remain active (Altmann 2005, Larkin & Kaye 2006). In docetaxel-refractory CRPC, ixabepilone and patupilone, as single agents, exhibit modest phase II clinical activity with PSA response rates around 20% (Rosenberg et al. 2007, Wilding et al. 2008, Hussain et al. 2009; Table 4). In combination with mitoxantrone in docetaxel-resistant CRPC, ixabepilone resulted in a PSA response rate of 31% (Rosenberg et al. 2009). A more promising phase II study of patupilone alone in 83 patients with docetaxel-resistant CRPC revealed a PSA response rate of 45% (Beardsley et al. 2009). While these agents are active in the taxane refractory setting, they have not yet entered phase III testing.
Conclusion
Prostate cancer represents a large burden of disease in our community and there are still limited therapeutic options available in the advanced castration-resistant setting. Docetaxel is the only cytotoxic agent that consistently improves survival; however, around half of patients will never respond to treatment, while all will eventually develop resistant disease. A greater understanding of resistance pathways is needed to both predict resistance early in the course of treatment and to ultimately manipulate this resistance and improve outcomes. While multiple aspects of resistance have been explored, few have been successfully manipulated in the clinical setting. Many agents are still in the early phases of clinical testing and with the large number of ongoing trials progress is anticipated. Until recently, identification of reliable resistance markers has been unproductive; however, recognition of MIC1 as an early marker of treatment response may be an early element in a new generation of biomarker discovery. At present, recognition of resistance occurs after at least 6 weeks of cytotoxic therapy (Tannock et al. 2004), by which time cumulative toxicity is significant. It is likely that resistance pathways differ among individual prostate cancers and that a set of multiple resistance markers encompassing various mechanisms of resistance will be required. Similarly, to combat resistance, effectively targeting multiple pathways simultaneously will almost certainly be necessary. Exploration of interactions among resistance pathways rather than their study in isolation may facilitate these goals.
Declaration of interest
The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.
Funding
This work was supported by a Pfizer Cancer Research Grant; the National Health and Medical Research Council; the Cancer Institute of New South Wales; the Cancer Council of New South Wales; Cancer Australia; and the RT Hall Estate.
References
AIHW (Australian Institute of Health and Welfare) and AACR (Australasian Association of Cancer Registries) 2008 Cancer in Australia: an overview. Cat. no. CAN42. Canberra, Australia:AIHW.
Altmann KH 2005 Recent developments in the chemical biology of epothilones. Current Pharmaceutical Design 11 1595–1613 doi:10.2174/1381612053764715.
Amantana A, London CA, Iversen PL & Devi GR 2004 X-linked inhibitor of apoptosis protein inhibition induces apoptosis and enhances chemotherapy sensitivity in human prostate cancer cells. Molecular Cancer Therapeutics 3 699–707.
Aragon-Ching JB & Dahut WL 2008 The role of angiogenesis inhibitors in prostate cancer. Cancer Journal 14 20–25 doi:10.1097/PPO.0b013e318161c014.
Araki S, Omori Y, Lyn D, Singh RK, Meinbach DM, Sandman Y, Lokeshwar VB & Lokeshwar BL 2007 Interleukin-8 is a molecular determinant of androgen independence and progression in prostate cancer. Cancer Research 67 6854–6862 doi:10.1158/0008-5472.CAN-07-1162.
Banerji U 2009 Heat shock protein 90 as a drug target: some like it hot. Clinical Cancer Research 15 9–14 doi:10.1158/1078-0432.CCR-08-0132.
Bauskin AR, Brown DA, Junankar S, Rasiah KK, Eggleton S, Hunter M, Liu T, Smith D, Kuffner T & Pankhurst GJ et al. 2005 The propeptide mediates formation of stromal stores of PROMIC-1: role in determining prostate cancer outcome. Cancer Research 65 2330–2336 doi:10.1158/0008-5472.CAN-04-3827.
Bauskin AR, Brown DA, Kuffner T, Johnen H, Luo XW, Hunter M & Breit SN 2006 Role of macrophage inhibitory cytokine-1 in tumorigenesis and diagnosis of cancer. Cancer Research 66 4983–4986 doi:10.1158/0008-5472.CAN-05-4067.
Beardsley EK, Saad F, Eigl B, Venner P, Hotte S, Winquist E, Ko YJ, Sridhar SS & Chi KN 2009 A phase II study of patupilone in patients (pts) with metastatic castration-resistant prostate cancer (CRPC) who have progressed after docetaxel. ASCO Annual meeting abstract. Journal of Clinical Oncology 27 (15S) abstract 5139.
Beer TM, Lalani AS, Lee S, Mori M, Eilers KM, Curd JG, Henner WD, Ryan CW, Venner P & Ruether JD et al. 2008 C-reactive protein as a prognostic marker for men with androgen-independent prostate cancer: results from the ASCENT trial. Cancer 112 2377–2383 doi:10.1002/cncr.23461.
Bellone M, Mondino A & Corti A 2008 Vascular targeting, chemotherapy and active immunotherapy: teaming up to attack cancer. Trends in Immunology 29 235–241 doi:10.1016/j.it.2008.02.003.
Bond GL, Hu W & Levine AJ 2005 MDM2 is a central node in the p53 pathway: 12 years and counting. Current Cancer Drug Targets 5 3–8.
Borgstrom P, Bourdon MA, Hillan KJ, Sriramarao P & Ferrara N 1998 Neutralizing anti-vascular endothelial growth factor antibody completely inhibits angiogenesis and growth of human prostate carcinoma micro tumors in vivo. Prostate 35 1–10.
Borsellino N, Belldegrun A & Bonavida B 1995 Endogenous interleukin 6 is a resistance factor for cis-diamminedichloroplatinum and etoposide-mediated cytotoxicity of human prostate carcinoma cell lines. Cancer Research 55 4633–4639.
Bradshaw DM & Arceci RJ 1998 Clinical relevance of transmembrane drug efflux as a mechanism of multidrug resistance. Journal of Clinical Oncology 16 3674–3690.
Brat DJ, Bellail AC & Van Meir EG 2005 The role of interleukin-8 and its receptors in gliomagenesis and tumoral angiogenesis. Neuro-Oncology 7 122–133 doi:10.1215/S1152851704001061.
Brown DA, Stephan C, Ward RL, Law M, Hunter M, Bauskin AR, Amin J, Jung K, Diamandis EP & Hampton GM et al. 2006 Measurement of serum levels of macrophage inhibitory cytokine 1 combined with prostate-specific antigen improves prostate cancer diagnosis. Clinical Cancer Research 12 89–96 doi:10.1158/1078-0432.CCR-05-1331.
Castellano DE, Gonzalez-Larriba J, Anton-Aparicio LM, Cassinello J, Grande Pulido E, Esteban E 2010 Clinical, serological, and radiographic benefit with sunitinib as a single agent in metastatic chemotherapy-resistant and hormone-refractory prostate cancer (HRPC) patients. In ASCO 2010 Genitourinary Cancers Symposium, abstract 229.
Cavarretta IT, Neuwirt H, Untergasser G, Moser PL, Zaki MH, Steiner H, Rumpold H, Fuchs D, Hobisch A & Nemeth JA et al. 2007 The antiapoptotic effect of IL-6 autocrine loop in a cellular model of advanced prostate cancer is mediated by Mcl-1. Oncogene 26 2822–2832 doi:10.1038/sj.onc.1210097.
Cetnar JP, Rosen MA, Vaughn DJ, Haas NB, Troxel AB, Song H, Adluru G, Flaherty KT, O'Dwyer PJ & Amaravadi RK 2009 Phase II study of sorafenib and docetaxel in men with metastatic castration resistant prostate cancer (mCRPC). ASCO Meeting. Journal of Clinical Oncology 27 abstract e16055.
Chan JM, Stampfer MJ, Ma J, Gann P, Gaziano JM, Pollak M & Giovannucci E 2002 Insulin-like growth factor-I (IGF-I) and IGF binding protein-3 as predictors of advanced-stage prostate cancer. Journal of National Cancer Institute 94 1099–1106 doi:10.1093/jnci/94.14.1099.
Chen T, Wang LH & Farrar WL 2000 Interleukin 6 activates androgen receptor-mediated gene expression through a signal transducer and activator of transcription 3-dependent pathway in LNCaP prostate cancer cells. Cancer Research 60 2132–2135.
Chi KN, Gleave ME, Klasa R, Murray N, Bryce C, Lopes de Menezes DE, D'Aloisio S & Tolcher AW 2001 A phase I dose-finding study of combined treatment with an antisense Bcl-2 oligonucleotide (Genasense) and mitoxantrone in patients with metastatic hormone-refractory prostate cancer. Clinical Cancer Research 7 3920–3927.
Chi KN, Eisenhauer E, Fazli L, Jones EC, Goldenberg SL, Powers J, Tu D & Gleave ME 2005a A phase I pharmacokinetic and pharmacodynamic study of OGX-011, a 2′-methoxyethyl antisense oligonucleotide to clusterin, in patients with localized prostate cancer. Journal of National Cancer Institute 97 1287–1296 doi:10.1093/jnci/dji252.
Chi KN, Eisenhauer E, Siu L, Hirte H, Hotte SJ, Chia S, Knox J, Guns E, Powers J & Gleave ME 2005b A phase I study of a second generation antisense oligonucleotide to clusterin (OGX-011) in combination with docetaxel: NCIC CTG IND.154. Journal of Clinical Oncology 23 (16S) abstract 3085.
Chi KN, Bjartell A, Dearnaley D, Saad F, Schröder FH, Sternberg C, Tombal B & Visakorpi T 2009a Castration-resistant prostate cancer: from new pathophysiology to new treatment targets. European Urology 56 594–605 doi:10.1016/j.eururo.2009.06.027.
Chi KN, Hotte SJ, Yu E, Tu D, Eigl B, Tannock I, Saad F, North S, Powers J & Eisenhauer E et al. 2009b Mature results of a randomized phase II study of OGX-011 in combination with docetaxel/prednisone versus docetaxel/prednisone in patients with metastatic castration-resistant prostate cancer. ASCO Annual meeting abstract. Journal of Clinical Oncology 27 (15S) abstract 5012.
Chiou SK, Jones MK & Tarnawski AS 2003 Survivin – an anti-apoptosis protein: its biological roles and implications for cancer and beyond. Medical Science Monitor 9 PI25–PI29.
Chopra DP, Menard RE, Januszewski J & Mattingly RR 2004 TNF-alpha-mediated apoptosis in normal human prostate epithelial cells and tumor cell lines. Cancer Letters 203 145–154 doi:10.1016/j.canlet.2003.09.016.
Chuang SE, Yeh PY, Lu YS, Lai GM, Liao CM, Gao M & Cheng AL 2002 Basal levels and patterns of anticancer drug-induced activation of nuclear factor-kappaB (NF-kappaB), and its attenuation by tamoxifen, dexamethasone, and curcumin in carcinoma cells. Biochemical Pharmacology 63 1709–1716.
Chung TD, Yu JJ, Spiotto MT, Bartkowski M & Simons JW 1999 Characterization of the role of IL-6 in the progression of prostate cancer. Prostate 38 199–207.
Culig Z, Oh SJ, Santer FR, Puhr M, Steiner H & Hobisch A 2009 Effects of sorafenib on proliferation of hormone-sensitive and hormone-insensitive prostate cancer cells. ASCO Meeting. Journal of Clinical Oncology 27 abstract e16092.
Cumashi A, Tinari N, Rossi C, Lattanzio R, Natoli C, Piantelli M & Iacobelli S 2008 Sunitinib malate (SU-11248) alone or in combination with low-dose docetaxel inhibits the growth of DU-145 prostate cancer xenografts. Cancer Letters 270 229–233 doi:10.1016/j.canlet.2008.05.007.
Dahut WL, Scripture CD, Posadas EM, Wu S, Arlen PM, Gulley JL, Wright J, Chen CC, Jones E & Figg WD 2006 Bony metastatic disease responses to sorafenib (BAY 43-9006) independent of PSA in patients with metastatic androgen independent prostate cancer. ASCO Meeting. Journal of Clinical Oncology 24 abstract 4506.
Dash R, Richards JE, Su ZZ, Bhutia SK, Azab B, Rahmani M, Dasmahapatra G, Yacoub A, Dent P & Dmitriev IP et al. 2010 Mechanism by which Mcl-1 regulates cancer-specific apoptosis triggered by mda-7/IL-24, an IL-10-related cytokine. Cancer Research 70 5034–5045 doi:10.1158/0008-5472.CAN-10-0563.
Dean E, Jodrell D, Connolly K, Danson S, Jolivet J, Durkin J, Morris S, Jowle D, Ward T & Cummings J et al. 2009 Phase I trial of AEG35156 administered as a 7-day and 3-day continuous intravenous infusion in patients with advanced refractory cancer. Journal of Clinical Oncology 27 1660–1666 doi:10.1200/JCO.2008.19.5677.
De Bono JS, Fizazi K, Flechon A, Heidenreich A, Voog E, Davis NB, Qi M, Bandekar R, Vermeulen J, Hudes GR 2010a Randomized phase II study of CNTO 328 (C), an anti-IL-6 monoclonal antibody, in combination with mitoxantrone (M) versus M alone in metastatic castration-resistant prostate cancer (CRPC). In ASCO 2010 Genitourinary Cancers Symposium, abstract 86.
De Bono JS, Oudard S, Ozguroglu M, Hansen S, Machiels JH, Shen L, Matthews P, Sartor AO & For the TROPIC Investigators 2010b Cabazitaxel or mitoxantrone with prednisone in patients with metastatic castration-resistant prostate cancer (mCRPC) previously treated with docetaxel: final results of a multinational phase III trial (TROPIC). Journal of Clinical Oncology 28 4508.
De Bono JS, Oudard S, Ozguroglu M, Hansen S, Machiels JP, Kocak I, Gravis G, Bodrogi I, Mackenzie MJ & Shen L et al. 2010c Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet 376 1147–1154 doi:10.1016/S0140-6736(10)61389-X.
De Marzo AM, Platz EA, Sutcliffe S, Xu J, Gronberg H, Drake CG, Nakai Y, Isaacs WB & Nelson WG 2007 Inflammation in prostate carcinogenesis. Nature Reviews Cancer 7 256–269 doi:10.1038/nrc2090.
Deveraux QL, Roy N, Stennicke HR, Van Arsdale T, Zhou Q, Srinivasula SM, Alnemri ES, Salvesen GS & Reed JC 1998 IAPs block apoptotic events induced by caspase-8 and cytochrome c by direct inhibition of distinct caspases. EMBO Journal 17 2215–2223 doi:10.1093/emboj/17.8.2215.
Di Lorenzo G, Figg WD, Fossa SD, Mirone V, Autorino R, Longo N, Imbimbo C, Perdona S, Giordano A & Giuliano M 2008 Combination of bevacizumab and docetaxel in docetaxel-pretreated hormone-refractory prostate cancer: a phase 2 study. European Urology 54 1089–1096 doi:10.1016/j.eururo.2008.01.082.
Domingo-Domenech J, Oliva C, Rovira A, Codony-Servat J, Bosch M, Filella X, Montagut C, Tapia M, Campas C & Dang L 2006 Interleukin 6, a nuclear factor-kappaB target, predicts resistance to docetaxel in hormone-independent prostate cancer and nuclear factor-kappaB inhibition by PS-1145 enhances docetaxel antitumor activity. Clinical Cancer Research 12 5578–5586 doi:10.1158/1078-0432.CCR-05-2767.
Domingo-Domenech J, Martinez-Fernandez A, Garcia-Albeniz X, Fernandez P, Filella X, Gascon P & Mellado B 2007 Correlation of serum interleukin-6 (IL-6) levels with immunohistochemical p65/nuclear factor-{kappa}B (NF-{kappa}B) expression and prediction of the clinical outcome of hormone-independent prostate cancer patients (HIPC) treated with docetaxel (D). ASCO Annual meeting abstract. Journal of Clinical Oncology 25 (18S) abstract 5075.
Dror Michaelson M, Regan MM, Oh WK, Kaufman DS, Olivier K, Michaelson SZ, Spicer B, Gurski C, Kantoff PW & Smith MR 2009 Phase II study of sunitinib in men with advanced prostate cancer. Annals of Oncology 20 913–920 doi:10.1093/annonc/mdp111.
Duckett CS, Nava VE, Gedrich RW, Clem RJ, Van Dongen JL, Gilfillan MC, Shiels H, Hardwick JM & Thompson CB 1996 A conserved family of cellular genes related to the baculovirus iap gene and encoding apoptosis inhibitors. EMBO Journal 15 2685–2694.
Epstein RJ 2007 VEGF signaling inhibitors: more pro-apoptotic than anti-angiogenic. Cancer Metastasis Reviews 26 443–452 doi:10.1007/s10555-007-9071-1.
Ferlay J Bray F, Pisani P, Parkin D 2004 GLOBOCAN 2002: cancer incidence, mortality and prevalence worldwide. IARC Cancer Base, vol 5.
Figg WD, Arlen P, Gulley J, Fernandez P, Noone M, Fedenko K, Hamilton M, Parker C, Kruger EA & Pluda J 2001a A randomized phase II trial of docetaxel (taxotere) plus thalidomide in androgen-independent prostate cancer. Seminars in Oncology 28 62–66.
Figg WD, Dahut W, Duray P, Hamilton M, Tompkins A, Steinberg SM, Jones E, Premkumar A, Linehan WM & Floeter MK et al. 2001b A randomized phase II trial of thalidomide, an angiogenesis inhibitor, in patients with androgen-independent prostate cancer. Clinical Cancer Research 7 1888–1893.
Figg WD, Retter A, Steinberg SM & Dahut WL 2005 In reply. Journal of Clinical Oncology 23 2113–2114.
Figueroa JA, De Raad S, Tadlock L, Speights VO & Rinehart JJ 1998 Differential expression of insulin-like growth factor binding proteins in high versus low Gleason score prostate cancer. Journal of Oncology 159 1379–1383.
Fox WD, Higgins B, Maiese KM, Drobnjak M, Cordon-Cardo C, Scher HI & Agus DB 2002 Antibody to vascular endothelial growth factor slows growth of an androgen-independent xenograft model of prostate cancer. Clinical Cancer Research 8 3226–3231.
Fracasso PM, Goldstein LJ, de Alwis DP, Rader JS, Arquette MA, Goodner SA, Wright LP, Fears CL, Gazak RJ & Andre VA et al. 2004 Phase I study of docetaxel in combination with the P-glycoprotein inhibitor, zosuquidar, in resistant malignancies. Clinical Cancer Research 10 7220–7228 doi:10.1158/1078-0432.CCR-04-0452.
Gabai VL, Budagova KR & Sherman MY 2005 Increased expression of the major heat shock protein Hsp72 in human prostate carcinoma cells is dispensable for their viability but confers resistance to a variety of anticancer agents. Oncogene 24 3328–3338 doi:10.1038/sj.onc.1208495.
Gelmon KA, Latreille J, Tolcher A, Genier L, Fisher B, Forand D, D'Aloisio S, Vernillet L, Daigneault L & Lebecq A et al. 2000 Phase I dose-finding study of a new taxane, RPR 109881A, administered as a one-hour intravenous infusion days 1 and 8 to patients with advanced solid tumors. J Clin Oncol 18 4098–4108.
George DJ, Halabi S, Shepard TF, Vogelzang NJ, Hayes DF, Small EJ & Kantoff PW 2001 Prognostic significance of plasma vascular endothelial growth factor levels in patients with hormone-refractory prostate cancer treated on Cancer and Leukemia Group B 9480. Clinical Cancer Research 7 1932–1936.
Gillison TL, Appleman LJ, Friedland DM, Evans TL, Lara PN, Gooding WE, Lenzner DE, Strausser HM, Gingrich JR & Chatta GS 2009 Docetaxel and imatinib every 21 days for castration resistant prostate cancer: a phase II trial. ASCO Annual meeting abstract. Journal of Clinical Oncology 27 (15S) abstract e16086.
Gleave M, Miyake H & Chi K 2005 Beyond simple castration: targeting the molecular basis of treatment resistance in advanced prostate cancer. Cancer Chemotherapy and Pharmacology 56 (Supplement 1) 47–57 doi:10.1007/s00280-005-0098-0.
Gomez-Pinillos A, Ballard H, Shelton G, Reilly MM, Chachoua A, Taneja S & Ferrari AC 2009 Sequential and intermittent docetaxel (D) and imatinib (Im) in hormone-refractory prostate cancer patients (NYU 04-47). ASCO Annual meeting abstract. Journal of Clinical Oncology 27 (15S) abstract e16108.
Gross ME, Soscia J, Sakowsky S, Castellanos O & Agus DB 2009 Phase I trial of RAD001 (R), bevacizumab (B), and docetaxel (D) for castration-resistant prostate cancer (CRPC). ASCO Annual meeting abstract. Journal of Clinical Oncology 27 (15S) abstract 5154.
Grunwald V, DeGraffenried L, Russel D, Friedrichs WE, Ray RB & Hidalgo M 2002 Inhibitors of mTOR reverse doxorubicin resistance conferred by PTEN status in prostate cancer cells. Cancer Research 62 6141–6145.
Guerin O, Formento P, Lo Nigro C, Hofman P, Fischel JL, Etienne-Grimaldi MC, Merlano M, Ferrero JM & Milano G 2008 Supra-additive antitumor effect of sunitinib malate (SU11248, Sutent) combined with docetaxel. A new therapeutic perspective in hormone refractory prostate cancer. Journal of Cancer Research and Clinical Oncology 134 51–57 doi:10.1007/s00432-007-0247-4.
Haldar S, Chintapalli J & Croce CM 1996 Taxol induces bcl-2 phosphorylation and death of prostate cancer cells. Cancer Research 56 1253–1255.
Haupt Y, Maya R, Kazaz A & Oren M 1997 Mdm2 promotes the rapid degradation of p53. Nature 387 296–299 doi:10.1038/387296a0.
Hayashi N, Asano K, Suzuki H, Yamamoto T, Tanigawa N, Egawa S & Manome Y 2005 Adenoviral infection of survivin antisense sensitizes prostate cancer cells to etoposide in vivo. Prostate 65 10–19 doi:10.1002/pros.20232.
Heldin CH, Rubin K, Pietras K & Ostman A 2004 High interstitial fluid pressure – an obstacle in cancer therapy. Nature Reviews Cancer 4 806–813 doi:10.1038/nrc1456.
Hobisch A, Eder IE, Putz T, Horninger W, Bartsch G, Klocker H & Culig Z 1998 Interleukin-6 regulates prostate-specific protein expression in prostate carcinoma cells by activation of the androgen receptor. Cancer Research 58 4640–4645.
Hofer MD, Fecko A, Shen R, Setlur SR, Pienta KG, Tomlins SA, Chinnaiyan AM & Rubin MA 2004 Expression of the platelet-derived growth factor receptor in prostate cancer and treatment implications with tyrosine kinase inhibitors. Neoplasia 6 503–512.
Honda R, Tanaka H & Yasuda H 1997 Oncoprotein MDM2 is a ubiquitin ligase E3 for tumor suppressor p53. FEBS Letters 420 25–27 doi:10.1016/S0014-5793(97)01480-4.
Hour T-C, Chen J, Huang C-Y, Guan J-Y, Lu S-H & Pu Y-S 2002 Curcumin enhances cytotoxicity of chemotherapeutic agents in prostate cancer cells by inducing p21(WAF1/CIP1) and C/EBPbeta expressions and suppressing NF-kappaB activation. Prostate 51 211–218 doi:10.1002/pros.10089.
Huang CY, Beer TM, Higano CS, True LD, Vessella R, Lange PH, Garzotto M & Nelson PS 2007 Molecular alterations in prostate carcinomas that associate with in vivo exposure to chemotherapy: identification of a cytoprotective mechanism involving growth differentiation factor 15. Clinical Cancer Research 13 5825–5833 doi:10.1158/1078-0432.CCR-07-1037.
Hudes G, Tagawa S, Whang Y, Qi M, Qin X, Puchalski T, Prabhakar U, O'Brien K & Eisenberger M 2009 A phase I study of CNTO328, an anti-interleukin (IL)-6 monoclonal antibody combined with docetaxel in subjects with metastatic castration-resistant prostate cancer (CRPC). Journal of Clinical Oncology 27 5063.
Hussain M, Tangen CM, Lara PN Jr, Vaishampayan UN, Petrylak DP, Colevas AD, Sakr WA & Crawford ED 2005 Ixabepilone (epothilone B analogue BMS-247550) is active in chemotherapynaive patients with hormone-refractory prostate cancer: a Southwest Oncology Group trial SO111. J Clin Oncol 23 8724–8729 doi:10.1200/JCO.2005.02.4448.
Hussain A, DiPaola RS, Baron AD, Higano CS, Tchekmedyian NS & Johri AR 2009 Phase II trial of weekly patupilone in patients with castration-resistant prostate cancer. Annals of Oncology 20 492–497 doi:10.1093/annonc/mdn665.
Isambert N, Freyer G, Zanetta S, Falandry C, Soussan Lazard K & Fumoleau PA 2008 A phase I dose escalation and pharmacokinetic (PK) study of intravenous aflibercept (VEGF trap) plus docetaxel (D) in patients (pts) with advanced solid tumors: preliminary results. Journal of Clinical Oncology 26 3599.
Jain RK 2005 Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 307 58–62 doi:10.1126/science.1104819.
Jemal A, Siegel R, Ward E, Hao Y, Xu J & Thun MJ 2009 Cancer statistics, 2009. CA: A Cancer Journal for Clinicians 59 225–249 doi:10.3322/caac.20006.
Kamath K, Wilson L, Cabral F & Jordan MA 2005 BetaIII-tubulin induces paclitaxel resistance in association with reduced effects on microtubule dynamic instability. Journal of Biological Chemistry 280 12902–12907 doi:10.1074/jbc.M414477200.
Kantoff PW, Halabi S, Conaway M, Picus J, Kirshner J, Hars V, Trump D, Winer EP & Vogelzang NJ 1999 Hydrocortisone with or without mitoxantrone in men with hormone-refractory prostate cancer: results of the cancer and leukemia group B 9182 study. Journal of Clinical Oncology 17 2506–2513.
Kelly WK, Halabi S, Carducci MA, George DJ, Mahoney JF, Stadler WM, Morris MJ, Kantoff PW, Monk JP III & Small EJ et al. 2010 A randomized, double-blind, placebo-controlled phase III trial comparing docetaxel, prednisone, and placebo with docetaxel, prednisone, and bevacizumab in men with metastatic castration-resistant prostate cancer (mCRPC): survival results of CALGB 90401. ASCO meeting abstract. Journal of Clinical Oncology 28 (18S) abstract LBA4511.
Kim SJ, Uehara H, Yazici S, Busby JE, Nakamura T, He J, Maya M, Logothetis C, Mathew P & Wang X et al. 2006 Targeting platelet-derived growth factor receptor on endothelial cells of multidrug-resistant prostate cancer. Journal of National Cancer Institute 98 783–793 doi:10.1093/jnci/djj211.
Krajewska M, Krajewski S, Epstein JI, Shabaik A, Sauvageot J, Song K, Kitada S & Reed JC 1996 Immunohistochemical analysis of bcl-2, bax, bcl-X, and mcl-1 expression in prostate cancers. American Journal of Pathology 148 1567–1576.
Kubler H, Van Randenborgh H, Treiber U, Lehmer A, Wagenpfeil S, Hartung R & Paul R 2004 Combined application of imatinib and standard chemotherapeutics results in additive cytotoxic effects in human prostate cancer cell lines. ASCO Annual meeting abstract. Journal of Clinical Oncology 22 (14S) abstract 4737.
LaCasse EC, Baird S, Korneluk RG & MacKenzie AE 1998 The inhibitors of apoptosis (IAPs) and their emerging role in cancer. Oncogene 17 3247–3259.
Lane DP 1992 Cancer. p53, guardian of the genome. Nature 358 15–16 doi:10.1038/358015a0.
Larkin JMG & Kaye SB 2006 Epothilones in the treatment of cancer. Expert Opinion on Investigational Drugs 15 691–702 doi:10.1517/13543784.15.6.691.
Lebedeva I, Rando R, Ojwang J, Cossum P & Stein CA 2000 Bcl-xL in prostate cancer cells: effects of overexpression and down-regulation on chemosensitivity. Cancer Research 60 6052–6060.
Lee JT Jr, Steelman LS & McCubrey JA 2004 Phosphatidylinositol 3′-kinase activation leads to multidrug resistance protein-1 expression and subsequent chemoresistance in advanced prostate cancer cells. Cancer Research 64 8397–8404 doi:10.1158/0008-5472.CAN-04-1612.
Lee JT, Lehmann BD, Terrian DM, Chappell WH, Stivala F, Libra M, Martelli AM, Steelman LS & McCubrey JA 2008 Targeting prostate cancer based on signal transduction and cell cycle pathways. Cell Cycle 7 1745–1762.
Leonetti C, Biroccio A, D'Angelo C, Semple SC, Scarsella M & Zupi G 2007 Therapeutic integration of c-myc and bcl-2 antisense molecules with docetaxel in a preclinical model of hormone-refractory prostate cancer. Prostate 67 1475–1485 doi:10.1002/pros.20636.
Leskov KS, Klokov DY, Li J, Kinsella TJ & Boothman DA 2003 Synthesis and functional analyses of nuclear clusterin, a cell death protein. Journal of Biological Chemistry 278 11590–11600 doi:10.1074/jbc.M209233200.
Li Y, Ahmed F, Ali S, Philip PA, Kucuk O & Sarkar FH 2005 Inactivation of nuclear factor kappaB by soy isoflavone genistein contributes to increased apoptosis induced by chemotherapeutic agents in human cancer cells. Cancer Research 65 6934–6942 doi:10.1158/0008-5472.CAN-04-4604.
Liston P, Roy N, Tamai K, Lefebvre C, Baird S, Cherton-Horvat G, Farahani R, McLean M, Ikeda JE & MacKenzie A et al. 1996 Suppression of apoptosis in mammalian cells by NAIP and a related family of IAP genes. Nature 379 349–353 doi:10.1038/379349a0.
Lokiec FM, Brain EG, Faivre S, Armand J-P, Gillotin C, Boissaye P, Marty M & Raymond E 2003 Docetaxel (D) and epirubicin (E) pharmacokinetic (PK) results in a phase I combination study with the novel oral p-glycoprotein (P-gp) inhibitor elacridar (GF120918) in patients (pts) with locally advanced or metastatic cancer. MDRF1002 study. Proceedings of the American Society of Clinical Oncology 22 abstract 614.
Luduena RF 1998 Multiple forms of tubulin: different gene products and covalent modifications. International Review of Cytology 178 207–275 doi:10.1016/S0074-7696(08)62138-5.
MacVicar GR, Greco A, Reeves J, Maleski J, Holmlund J & Leopold L 2009 An open-label, multicenter, phase I/II study of AT-101 in combination with docetaxel (D) and prednisone (P) in men with castrate-resistant prostate cancer (CRPC). Journal of Clinical Oncology 27 5062.
Makarovskiy AN, Siryaporn E, Hixson DC & Akerley W 2002 Survival of docetaxel-resistant prostate cancer cells in vitro depends on phenotype alterations and continuity of drug exposure. Cellular and Molecular Life Sciences 59 1198–1211 doi:10.1007/s00018-002-8498-3.
Marshall J, Chen H, Yang D, Figueira M, Bouker KB, Ling Y, Lippman M, Frankel SR & Hayes DF 2004 A phase I trial of a Bcl-2 antisense (G3139) and weekly docetaxel in patients with advanced breast cancer and other solid tumors. Annals of Oncology 15 1274–1283 doi:10.1093/annonc/mdh317.
Mathew P, Thall PF, Johnson MM, Oh WK, Meluch AA, Morris MJ, Troncoso P, Bucana CD, Fidler IJ & Logothetis CJ 2006 Preliminary results of a randomized placebo-controlled double-blind trial of weekly docetaxel combined with imatinib in men with metastatic androgen-independent prostate cancer (AIPC) and bone metastases (BM). ASCO Annual meeting abstract. Journal of Clinical Oncology 24 (18S) abstract 4562.
Mathew P, Thall PF, Wen S, Bucana C, Jones D, Horne E, Oh WK, Morris MJ, Lee YC & Logothetis CJ et al. 2008 Dynamic change in phosphorylated platelet-derived growth factor receptor in peripheral blood leukocytes following docetaxel therapy predicts progression-free and overall survival in prostate cancer. British Journal of Cancer 99 1426–1432 doi:10.1038/sj.bjc.6604706.
Maxwell PJ, Gallagher R, Seaton A, Wilson C, Scullin P, Pettigrew J, Stratford IJ, Williams KJ, Johnston PG & Waugh DJ 2007 HIF-1 and NF-kappaB-mediated upregulation of CXCR1 and CXCR2 expression promotes cell survival in hypoxic prostate cancer cells. Oncogene 26 7333–7345 doi:10.1038/sj.onc.1210536.
McDonnell TJ, Troncoso P, Brisbay SM, Logothetis C, Chung LW, Hsieh JT, Tu SM & Campbell ML 1992 Expression of the protooncogene bcl-2 in the prostate and its association with emergence of androgen-independent prostate cancer. Cancer Research 52 6940–6944.
Melnyk O, Zimmerman M, Kim KJ & Shuman M 1999 Neutralizing anti-vascular endothelial growth factor antibody inhibits further growth of established prostate cancer and metastases in a pre-clinical model. Journal of Urology 161 960–963.
Meric-Bernstam F & Gonzalez-Angulo AM 2009 Targeting the mTOR signaling network for cancer therapy. Journal of Clinical Oncology 27 2278–2287 doi:10.1200/JCO.2008.20.0766.
Mita AC, Denis LJ, Rowinsky EK, Debono JS, Goetz AD, Ochoa L, Forouzesh B, Beeram M, Patnaik A & Molpus K et al. 2009 Phase I and pharmacokinetic study of XRP6258 (RPR 116258A), a novel taxane, administered as a 1-hour infusion every 3 weeks in patients with advanced solid tumors. Clinical Cancer Research 15 723–730 doi:10.1158/1078-0432.CCR-08-0596.
Miyake H, Tolcher A & Gleave ME 2000a Chemosensitization and delayed androgen-independent recurrence of prostate cancer with the use of antisense Bcl-2 oligodeoxynucleotides. Journal of National Cancer Institute 92 34–41 doi:10.1093/jnci/92.1.34.
Miyake H, Chi KN & Gleave ME 2000b Antisense TRPM-2 oligodeoxynucleotides chemosensitize human androgen-independent PC-3 prostate cancer cells both in vitro and in vivo. Clinical Cancer Research 6 1655–1663.
Miyake H, Monia BP & Gleave ME 2000c Inhibition of progression to androgen-independence by combined adjuvant treatment with antisense BCL-XL and antisense Bcl-2 oligonucleotides plus taxol after castration in the Shionogi tumor model. International Journal of Cancer 86 855–862.
Miyake H, Nelson C, Rennie PS & Gleave ME 2000d Acquisition of chemoresistant phenotype by overexpression of the antiapoptotic gene testosterone-repressed prostate message-2 in prostate cancer xenograft models. Cancer Research 60 2547–2554.
Miyake H, Nelson C, Rennie PS & Gleave ME 2000e Overexpression of insulin-like growth factor binding protein-5 helps accelerate progression to androgen-independence in the human prostate LNCaP tumor model through activation of phosphatidylinositol 3′-kinase pathway. Endocrinology 141 2257–2265 doi:10.1210/en.141.6.2257.
Mizutani K, Matsumoto K, Hasegawa N, Deguchi T & Nozawa Y 2006 Expression of clusterin, XIAP and survivin, and their changes by camptothecin (CPT) treatment in CPT-resistant PC-3 and CPT-sensitive LNCaP cells. Experimental Oncology 28 209–215.
Momand J, Jung D, Wilczynski S & Niland J 1998 The MDM2 gene amplification database. Nucleic Acids Research 26 3453–3459.
Montgomery RB, Bonham M, Nelson PS, Grim J, Makary E, Vessella R & Stahl WL 2005 Estrogen effects on tubulin expression and taxane mediated cytotoxicity in prostate cancer cells. Prostate 65 141–150 doi:10.1002/pros.20246.
Montgomery B, Lin D, Ryan C, Garzotto M & Beer TM 2006 Diethylstilbestrol and docetaxel: a phase II study in patients with metastatic, androgen independent prostate cancer. ASCO Annual meeting abstract. Journal of Clinical Oncology 24 (18S) abstract 4627.
Morris MJ, Tong WP, Cordon-Cardo C, Drobnjak M, Kelly WK, Slovin SF, Terry KL, Siedlecki K, Swanson P & Rafi M et al. 2002 Phase I trial of BCL-2 antisense oligonucleotide (G3139) administered by continuous intravenous infusion in patients with advanced cancer. Clinical Cancer Research 8 679–683.
Morris MJ, Cordon-Cardo C, Kelly WK, Slovin SF, Siedlecki K, Regan KP, DiPaola RS, Rafi M, Rosen N & Scher HI 2005 Safety and biologic activity of intravenous BCL-2 antisense oligonucleotide (G3139) and taxane chemotherapy in patients with advanced cancer. Applied Immunohistochemistry & Molecular Morphology 13 6–13.
Moss RA, Shelton G, Melia J, Mohike SG, Petrylak DP 2007 A phase I open-label study using lenalidomide and docetaxel in androgen independent prostate cancer (AIPC). ASCO Prostate Cancer Symposium. abstract 89.
Musquire LA, Ramalingam S, Egorin MJ, Sikorski RP, Chatta GS, Ramanathan RK, Friedland D, Potter D, Ivy SP & Belani CP 2007 Phase I and pharmacokinetic (PK) study of 17-allylamino-17 demethoxygeldanamycin (17-AAG) in combination with weekly paclitaxel for advanced solid malignancies. ASCO Annual meeting abstract. Journal of Clinical Oncology 25 (18S) abstract 14028.
Nabhan C, Tolzien K, Lestingi TM, Kelby SK, Galvez AG, Bitran JD 2010 Activity of sorafenib (SOR) in chemotherapy-failure castration-resistant prostate cancer (CRPC). In ASCO Genitourinary Cancers Symposium, abstract 233.
Nakahara T, Takeuchi M, Kinoyama I, Minematsu T, Shirasuna K, Matsuhisa A, Kita A, Tominaga F, Yamanaka K & Kudoh M et al. 2007 YM155, a novel small-molecule survivin suppressant, induces regression of established human hormone-refractory prostate tumor xenografts. Cancer Research 67 8014–8021 doi:10.1158/0008-5472.CAN-07-1343.
Newman RA, Yang J, Raymond M, Finlay V, Cabral F, Vourloumis D, Stephens LC, Troncoso P, Wu X & Logothetis CJ et al. 2001 Antitumor efficacy of 26-fluoroepothilone B against human prostate cancer xenografts. Cancer Chemotherapy and Pharmacology 48 319–326.
Ning YM, Arlen PM, Gulley JL, Stein WD, Fojo AT, Latham L, Wright JJ, Parnes H, Figg WD & Dahut WL 2008 Phase II trial of thalidomide (T), bevacizumab (Bv), and docetaxel (Doc) in patients (pts) with metastatic castration-refractory prostate cancer (mCRPC). ASCO Annual meeting abstract. Journal of Clinical Oncology 26 (15S) abstract 5000.
Nomura T, Mimata H, Takeuchi Y, Yamamoto H, Miyamoto E & Nomura Y 2003 The X-linked inhibitor of apoptosis protein inhibits taxol-induced apoptosis in LNCaP cells. Urological Research 31 37–44 doi:10.1007/s00240-003-0300-y.
Oh WK, Stadler WM, Srinivas S, Chu F, Bubley G, Quigley M, Goddard J, Dunbar J, Grayzel D, Ross RW 2009 A single arm phase II trial of IPI-504 in patients with castration resistant prostate cancer (CRPC). In ASCO Genitourinary Cancers Symposium. abstract 219.
Patterson SG, Wei S, Chen X, Sallman DA, Gilvary DL, Zhong B, Pow-Sang J, Yeatman T & Djeu JY 2006 Novel role of stat1 in the development of docetaxel resistance in prostate tumor cells. Oncogene 25 6113–6122 doi:10.1038/sj.onc.1209632.
Paule B, Terry S, Kheuang L, Soyeux P, Vacherot F & de la Taille A 2007 The NF-kappaB/IL-6 pathway in metastatic androgen-independent prostate cancer: new therapeutic approaches? World Journal of Urology 25 477–489 doi:10.1007/s00345-007-0175-6.
Petrylak DP, Tangen CM, Hussain MH, Lara PN Jr, Jones JA, Taplin ME, Burch PA, Berry D, Moinpour C & Kohli M et al. 2004 Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer. New England Journal of Medicine 351 1513–1520 doi:10.1056/NEJMoa041318.
Pfeil K, Eder IE, Putz T, Ramoner R, Culig Z, Ueberall F, Bartsch G & Klocker H 2004 Long-term androgen-ablation causes increased resistance to PI3K/Akt pathway inhibition in prostate cancer cells. Prostate 58 259–268 doi:10.1002/pros.10332.
Picus J, Halabi S, Rini B, Vogelzang N, Whang Y, Kaplan E, Kelly W & Small E 2003 The use of bevacizumab (B) with docetaxel (D) and estramustine (E) in hormone refractory prostate cancer (HRPC): initial results of CALGB 90006. Proceedings of the American Society of Clinical Oncology 22 abstract 1578.
Pinski JK, Goldman B, Dorff T, Mack P, Lara P Jr, van Veldhuizen P, Quinn D, Hussain MH & Thompson IM 2009 SWOG S0354: a phase II trial of CNTO328, a monoclonal antibody against interleukin-6 (IL-6), in chemotherapy pretreated patients (pts) with castration-resistant prostate cancer (CRPC). ASCO Meeting. Journal of Clinical Oncology 27 abstract 5143.
Poiesz B, Reeves J, McNulty W, Maleski J, Holmlund J & Leopold L 2009 Preliminary report of an open-label, multicenter, phase I/II study of AT-101 in combination with docetaxel (D) and prednisone (P) in men with docetaxel refractory prostate cancer. Journal of Clinical Oncology 27 5145.
Presta LG, Chen H, O'Connor SJ, Chisholm V, Meng YG, Krummen L, Winkler M & Ferrara N 1997 Humanization of an anti-vascular endothelial growth factor monoclonal antibody for the therapy of solid tumors and other disorders. Cancer Research 57 4593–4599.
Pu YS, Hour TC, Chuang SE, Cheng AL, Lai MK & Kuo ML 2004 Interleukin-6 is responsible for drug resistance and anti-apoptotic effects in prostatic cancer cells. Prostate 60 120–129 doi:10.1002/pros.20057.
Raffo AJ, Perlman H, Chen MW, Day ML, Streitman JS & Buttyan R 1995 Overexpression of bcl-2 protects prostate cancer cells from apoptosis in vitro and confers resistance to androgen depletion in vivo. Cancer Research 55 4438–4445.
Rago RP, Einstein A Jr, Lush R, Beer TM, Ko Y-J, Henner WD, Bubley G, Merica EA, Garg V & Ette E et al. 2003 Safety and efficacy of the MDR inhibitor Incel (biricodar, VX-710) in combination with mitoxantrone and prednisone in hormone-refractory prostate cancer. Cancer Chemotherapy and Pharmacology 51 297–305 doi:10.1007/s00280-003-0573-4.
Rahman KM, Banerjee S, Ali S, Ahmad A, Wang Z, Kong D & Sakr WA 2009 3,3′-Diindolylmethane enhances taxotere-induced apoptosis in hormone-refractory prostate cancer cells through survivin down-regulation. Cancer Research 69 4468–4475 doi:10.1158/0008-5472.CAN-08-4423.
Ramalingam SS, Egorin MJ, Ramanathan RK, Remick SC, Sikorski RP, Lagattuta TF, Chatta GS, Friedland DM, Stoller RG & Potter DM et al. 2008 A phase I study of 17-allylamino-17-demethoxygeldanamycin combined with paclitaxel in patients with advanced solid malignancies. Clinical Cancer Research 14 3456–3461 doi:10.1158/1078-0432.CCR-07-5088.
Ranganathan S, Benetatos CA, Colarusso PJ, Dexter DW & Hudes GR 1998 Altered beta-tubulin isotype expression in paclitaxel-resistant human prostate carcinoma cells. British Journal of Cancer 77 562–566.
Reese DM, Fratesi P, Corry M, Novotny W, Holmgren E & Small EJ 2001 A phase II trial of humanized anti-vascular endothelial growth factor antibody for the treatment of androgen-independent prostate cancer. The Prostate Journal 3 65–70.
Ren A, Yan G, You B & Sun J 2008 Down-regulation of mammalian sterile 20-like kinase 1 by heat shock protein 70 mediates cisplatin resistance in prostate cancer cells. Cancer Research 68 2266–2274 doi:10.1158/0008-5472.CAN-07-6248.
Rocchi P, So A, Kojima S, Signaevsky M, Beraldi E, Fazli L, Hurtado-Coll A, Yamanaka K & Gleave M 2004 Heat shock protein 27 increases after androgen ablation and plays a cytoprotective role in hormone-refractory prostate cancer. Cancer Research 64 6595–6602 doi:10.1158/0008-5472.CAN-03-3998.
Rosenberg JE, Weinberg VK, Kelly WK, Michaelson D, Hussain MH, Wilding G, Gross M, Hutcheon D & Small EJ 2007 Activity of second-line chemotherapy in docetaxel-refractory hormone-refractory prostate cancer patients: randomized phase 2 study of ixabepilone or mitoxantrone and prednisone. Cancer 110 556–563 doi:10.1002/cncr.22811.
Rosenberg JE, Ryan CJ, Weinberg VK, Smith DC, Hussain M, Beer TM, Ryan CW, Mathew P, Pagliaro LC & Harzstark AL et al. 2009 Phase I study of ixabepilone, mitoxantrone, and prednisone in patients with metastatic castration-resistant prostate cancer previously treated with docetaxel-based therapy: a study of the Department of Defense Prostate Cancer Clinical Trials Consortium. Journal of Clinical Oncology 27 2772–2778 doi:10.1200/JCO.2008.19.8002.
Saad F, Hotte SJ, North SA, Eigl BJ, Chi KN, Czaykowski P, Polllak M, Wood L & Winquist E 2008 A phase II randomized study of custirsen (OGX-011) combination therapy in patients with poor-risk hormone refractory prostate cancer (HRPC) who relapsed on or within six months of 1st-line docetaxel therapy. Journal of Clinical Oncology 26 5002.
Sanchez C, Mendoza P, Contreras HR, Vergara J, McCubrey JA, Huidobro C & Castellon EA 2009 Expression of multidrug resistance proteins in prostate cancer is related with cell sensitivity to chemotherapeutic drugs. Prostate 69 1448–1459 doi:10.1002/pros.20991.
Saporita AJ, Ai J & Wang Z 2007 The Hsp90 inhibitor, 17-AAG, prevents the ligand-independent nuclear localization of androgen receptor in refractory prostate cancer cells. Prostate 67 509–520 doi:10.1002/pros.20541.
Satoh T, Okamoto I, Miyazaki M, Morinaga R, Tsuya A, Hasegawa Y, Terashima M, Ueda S, Fukuoka M & Ariyoshi Y et al. 2009 Phase I study of YM155, a novel survivin suppressant, in patients with advanced solid tumors. Clinical Cancer Research 15 3872–3880 doi:10.1158/1078-0432.CCR-08-1946.
Shaikh IA, Brown I, Schofield AC, Wahle KW & Heys SD 2008 Docosahexaenoic acid enhances the efficacy of docetaxel in prostate cancer cells by modulation of apoptosis: the role of genes associated with the NF-kappaB pathway. Prostate 68 1635–1646 doi:10.1002/pros.20830.
Singh S & Aggarwal BB 1995 Activation of transcription factor NF-kappa B is suppressed by curcumin (diferuloylmethane) [corrected]. Journal of Biological Chemistry 270 24995–25000.
Sissung TM, Baum CE, Deeken J, Price DK, Aragon-Ching J, Steinberg SM, Dahut W, Sparreboom A & Figg WD 2008 ABCB1 genetic variation influences the toxicity and clinical outcome of patients with androgen-independent prostate cancer treated with docetaxel. Clinical Cancer Research 14 4543–4549 doi:10.1158/1078-0432.CCR-07-4230.
Smith PC & Keller ET 2001 Anti-interleukin-6 monoclonal antibody induces regression of human prostate cancer xenografts in nude mice. Prostate 48 47–53 doi:10.1002/pros.1080.
Solit DB, Egorin M, Kopil C, Delacruz A, Shaffer D, Slovin S, Morris M, Kelly WK, Rosen N & Scher H 2005 Phase 1 pharmacokinetic and pharmacodynamic trial of docetaxel and 17AAG (17-allylamino-17-demethoxygeldanamycin). ASCO Annual meeting abstract. Journal of Clinical Oncology 23 (16S) abstract 3051.
Sowery RD, Hadaschik BA, So AI, Zoubeidi A, Fazli L, Hurtado-Coll A & Gleave ME 2008 Clusterin knockdown using the antisense oligonucleotide OGX-011 re-sensitizes docetaxel-refractory prostate cancer PC-3 cells to chemotherapy. BJU International 102 389–397 doi:10.1111/j.1464-410X.2008.07618.x.
Springate CM, Jackson JK, Gleave ME & Burt HM 2005 Efficacy of an intratumoral controlled release formulation of clusterin antisense oligonucleotide complexed with chitosan containing paclitaxel or docetaxel in prostate cancer xenograft models. Cancer Chemotherapy and Pharmacology 56 239–247 doi:10.1007/s00280-004-0997-5.
Srinivasan S, Kumar R, Koduru S, Chandramouli A & Damodaran C 2010 Inhibiting TNF-mediated signaling: a novel therapeutic paradigm for androgen independent prostate cancer. Apoptosis 15 153–161 doi:10.1007/s10495-009-0416-9.
Stephanou A & Latchman DS 2003 STAT-1: a novel regulator of apoptosis. International Journal of Experimental Pathology 84 239–244 doi:10.1111/j.0959-9673.2003.00363.x.
Sternberg CN, Dumez H, Van Poppel H, Skoneczna I, Sella A, Daugaard G, Gil T, Graham J, Carpentier P & Calabro F et al. 2009a Docetaxel plus oblimersen sodium (bcl-2 antisense oligonucleotide): an EORTC multicenter, randomized phase II study in patients with castration-resistant prostate cancer. Annals of Oncology 20 1264–1269 doi:10.1093/annonc/mdn784.
Sternberg CN, Petrylak DP, Sartor O, Witjes JA, Demkow T, Ferrero JM, Eymard JC, Falcon S, Calabro F & James N et al. 2009b Multinational, double-blind, phase III study of prednisone and either satraplatin or placebo in patients with castrate-refractory prostate cancer progressing after prior chemotherapy: the SPARC trial. Journal of Clinical Oncology 27 5431–5438 doi:10.1200/JCO.2008.20.1228.
Sullivan GF, Yang JM, Vassil A, Yang J, Bash-Babula J & Hait WN 2000 Regulation of expression of the multidrug resistance protein MRP1 by p53 in human prostate cancer cells. Journal of Clinical Investigation 105 1261–1267 doi:10.1172/JCI9290.
Sweeney CJ, Miller KD, Sissons SE, Nozaki S, Heilman DK, Shen J & Sledge GW Jr 2001 The antiangiogenic property of docetaxel is synergistic with a recombinant humanized monoclonal antibody against vascular endothelial growth factor or 2-methoxyestradiol but antagonized by endothelial growth factors. Cancer Research 61 3369–3372.
Sweeney C, Li L, Shanmugam R, Bhat-Nakshatri P, Jayaprakasan V, Baldridge LA, Gardner T, Smith M, Nakshatri H & Cheng L 2004 Nuclear factor-kappaB is constitutively activated in prostate cancer in vitro and is overexpressed in prostatic intraepithelial neoplasia and adenocarcinoma of the prostate. Clinical Cancer Research 10 5501–5507.
Takeda M, Mizokami A, Mamiya K, Li YQ, Zhang J, Keller ET & Namiki M 2007 The establishment of two paclitaxel-resistant prostate cancer cell lines and the mechanisms of paclitaxel resistance with two cell lines. Prostate 67 955–967 doi:10.1002/pros.20581.
Talbot DC, Davies J, Callies S, Andre V, Lahn M, Ang J, De Bono JS & Ranson M 2008 First human dose study evaluating safety and pharmacokinetics of LY2181308, an antisense oligonucleotide designed to inhibit survivin. ASCO Annual meeting abstract. Journal of Clinical Oncology 26 (15S) abstract 3518.
Tannock IF, Osoba D, Stockler MR, Ernst DS, Neville AJ, Moore MJ, Armitage GR, Wilson JJ, Venner PM & Coppin CM et al. 1996 Chemotherapy with mitoxantrone plus prednisone or prednisone alone for symptomatic hormone-resistant prostate cancer: a Canadian randomized trial with palliative end points. Journal of Clinical Oncology 14 1756–1764.
Tannock IF, de Wit R, Berry WR, Horti J, Pluzanska A, Chi KN, Oudard S, Theodore C, James ND & Turesson I et al. 2004 Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. New England Journal of Medicine 351 1502–1512 doi:10.1056/NEJMoa040720.
Tohnya TM, Gulley J, Arlen PM, Sparreboom A, Venitz J, Parker C, Fedenko K, Parnes H, Figg WD & Dahut WL 2006 Phase I study of lenalidomide, a novel thalidomide analog, in patients with refractory metastatic cancer. ASCO Annual meeting abstract. Journal of Clinical Oncology 24 (18S) abstract 13038.
Tolcher AW, Kuhn J, Schwartz G, Patnaik A, Hammond LA, Thompson I, Fingert H, Bushnell D, Malik S & Kreisberg J et al. 2004 A Phase I pharmacokinetic and biological correlative study of oblimersen sodium (genasense, g3139), an antisense oligonucleotide to the bcl-2 mRNA, and of docetaxel in patients with hormone-refractory prostate cancer. Clinical Cancer Research 10 5048–5057.
Tolcher AW, Mita A, Lewis LD, Garrett CR, Till E, Daud AI, Patnaik A, Papadopoulos K, Takimoto C & Bartels P et al. 2008 Phase I and pharmacokinetic study of YM155, a small-molecule inhibitor of survivin. Journal of Clinical Oncology 26 5198–5203 doi:10.1200/JCO.2008.17.2064.
Visa L, Pineda E, Farrus B, Codony-Servat J, Filella X, Albiol S, Martinez A, Domingo-Domench JM, Gascon P & Mellado B 2009 Correlation of serum interleukin-6 (IL-6) levels and clinical outcome in hormone-independent (HI) prostate cancer (PC) patients (PTS) treated with docetaxel. ASCO Annual meeting abstract. Journal of Clinical Oncology 27 (15S) abstract e16044.
Wallner L, Dai J, Escara-Wilke J, Zhang J, Yao Z, Lu Y, Trikha M, Nemeth JA, Zaki MH & Keller ET 2006 Inhibition of interleukin-6 with CNTO328, an anti-interleukin-6 monoclonal antibody, inhibits conversion of androgen-dependent prostate cancer to an androgen-independent phenotype in orchiectomized mice. Cancer Research 66 3087–3095 doi:10.1158/0008-5472.CAN-05-3447.
Wang H, Yu D, Agrawal S & Zhang R 2003 Experimental therapy of human prostate cancer by inhibiting MDM2 expression with novel mixed-backbone antisense oligonucleotides: in vitro and in vivo activities and mechanisms. Prostate 54 194–205 doi:10.1002/pros.10187.
Wilding G, Chen Y, DiPaola RP, Carducci MA & Liu G 2008 E3803: updated results on phase II study of a weekly schedule of BMS-247550 for patients with castrate refractory prostate cancer (CRPC). ASCO Annual meeting abstract. Journal of Clinical Oncology 26 (15S) abstract 5070.
Wilson MR & Easterbrook-Smith SB 2000 Clusterin is a secreted mammalian chaperone. Trends in Biochemical Science 25 95