Abstract
For men with castration-resistant prostate cancer (CRPC), androgen-deprivation therapy (ADT) often becomes ineffective requiring the addition of docetaxel, a proven effective chemotherapy option. Tumor-associated macrophages (TAMs) are known to provide protumorigenic influences that contribute to treatment failure. In this study, we examined the contribution of TAMs to docetaxel treatment. An increased infiltration of macrophages in CRPC tumors was observed after treatment with docetaxel. Prostate cancer cells treated with docetaxel released more macrophage colony-stimulating factor (M-CSF-1 or CSF-1), IL-10 and other factors, which can recruit and modulate circulating monocytes to promote their protumorigenic functions. Inhibition of CSF-1 receptor kinase signaling with a small molecule antagonist (PLX3397) in CRPC models significantly reduces the infiltration of TAMs and their influences. As such, the addition of PLX3397 to docetaxel treatment resulted in a more durable tumor growth suppression than docetaxel alone. This study reveals a rational strategy to abrogate the influences of TAMs and extend the treatment response to docetaxel in CRPC.
Introduction
Prostate cancer (PCa) is the second most common cancer in men after skin cancer, as one out of seven men will be diagnosed with this disease in the United States by 2017 (Siegel et al. 2017). It is estimated that 161,000 newly diagnosed cases and 27,000 deaths will be attributed to this disease in 2017 (Siegel et al. 2017). A great majority of PCa patients, 70–80%, present with localized, organ-confined disease and their outcome is very favorable, having 10-year survival rate above 95%. However, 20–30% of patients will present with characteristics of high risk, advanced disease such as high Gleason grade or distant metastases. In these cases, the 5-year survival rate drops precipitously to about 30% (Siegel et al. 2017).
For PCa patients with advanced disease, androgen-deprivation therapy (ADT) is the first line of treatment, developed by Dr Huggins more than 75 years ago to deplete androgen, a key growth factor for prostate cancer cells (Esch et al. 2014). Over the years, effective strategies of ADT include the depletion of the body’s source of androgen by inhibiting androgen biosynthesis pathways and by blocking the activation of androgen receptor (AR) (Merseburger et al. 2015). Abiraterone and enzalutamide are two newly approved potent ADT agents that inhibit CYP17A1 androgen synthetic enzyme and AR, respectively (de Bono et al. 2011, Scher et al. 2012). Both agents are effective in prolonging the survival of castration-resistant prostate cancer (CRPC) patients who had progressed on first-line ADT (Ryan et al. 2015). However, a significant proportion of CRPC patients either do not respond to either abiraterone or enzalutamide, or initially respond but subsequently progress on treatment (Silberstein et al. 2016). Potential mechanisms of resistance include AR mutations, amplification and splice variant (Antonarakis et al. 2014, Azad et al. 2015, Romanel et al. 2015).
Docetaxel has been established as the standard first-line chemotherapy agent to treat CRPC since 2004. It was approved by FDA for this purpose as several large clinical trials showed docetaxel containing regimens provided survival benefits over other chemotherapies for CRPC patients (Petrylak et al. 2004, Tannock et al. 2004, Sweeney et al. 2015). Belonging to the taxane family, docetaxel was initially postulated to suppress prostate cancer growth by interfering with microtubule function (Petrylak 2003). However, subsequent research supported that the therapeutic activity of taxanes in prostate cancer could arise from its interference with androgen signaling via the nuclear translocation process (Gan et al. 2009).
Given taxane-based chemotherapy is one of a few effective treatments for CRPC, we investigate a rational combination regimen to improve its therapeutic efficacy. Recent findings from our group and others showed that tumor-associated macrophages (TAMs) contribute significantly to treatment failure in PCa and other solid cancers via their wound-healing and protumorigenic functions (Xu et al. 2013, Escamilla et al. 2015, Brown et al. 2017). In this study, we employed a small-molecule CSF1R kinase inhibitor (CSF-1Ri), PLX3397, to block TAMs in CRPC models. In combination with ADT and docetaxel, PLX3397 was able to significantly reduce the number of infiltrating TAMs and lower their protumorigenic influences. We showed that the addition of PLX3397 extended the therapeutic response to ADT and docetaxel in CRPC models.
Materials and methods
Cell culture and drugs
The murine macrophage RAW264.7 (RAW) cells (ATCC) and MyC-CaP cells (a kind gift from Dr. Charles Sawyers, Memorial Sloan Kettering New York) were cultured with DMEM (high glucose) while PC3 (ATCC), CWR22Rv2 (a kind gift from Dr. David Agus, Cedars-Sinai Medical Center) and LNCap-C4-2 (C4-2) cells (ATCC) were cultured in RPMI-1640. Both media were supplemented with 10% fetal bovine serum (FBS), 100 U/mL penicillin and 100 μg/mL streptomycin. PLX3397, 5-[(5-chloro-1H-pyrrolo[2,3-b]pyridin-3-yl)methyl]-N-[[6-(trifluoromethyl)-3-pyridyl]methyl]pyridin-2-amine was synthesized at Plexxikon Inc. The detailed synthetic procedure is shown by Tap et al. (2015).
Transwell coculture and migration assay
In coculture assay, 1.0 × 106 RAW macrophages were seeded in transwell inserts with membrane pore size at 4 μm (BD Falcon) in media supplemented with 2 μM PLX3397, 1 µM GW2580 or DMSO vehicle. The chamber was inserted in a 6-well plate with conditioned media from Myc-Cap, PC3, CWR and C4-2 cells treated with docetaxel (100 nM for MyC-CaP, 5 nM for CWR22Rv1, 30 nM for PC3 and 2 nM for C4-2 cells) or DMSO. Total RNA was extracted from tumor cells after 48 h and analyzed by RT-PCR. The methods for RT-PCR is described in Supplementary data (see section on supplementary data given at the end of this article) and primers are listed in Supplementary Table 1.
In migration assay, 1.0 × 105 RAW cells were seeded in transwell inserts with membrane pore size at 8 μm assembled in 24-well plates. The number of migrated cells was evaluated after 6 h of incubation at 37°C, and then treated with 3% paraformaldehyde (PFA) and stained with 0.1% (w/v) crystal violet solution. Random 10 fields/well at 4× magnification were sampled and quantified with ImageJ2.
ELISA assay
1.0 × 106 MyC-CaP, PC3, CWR and C4-2 cells were cocultured with or without RAW cells as mentioned earlier, with or without Docetaxel or PLX3397 at tumor cells’ IC50 or IC10 concentrations.
Supernatant of all cell culture media were harvested after 48 h. 96-well Nunc MaxiSorp Plates (Cat#44-2404-21, Thermo Scientific) were coated with the anti-M-CSF antibody (1:300, Cat#sc-365779, Santa Cruz Biotech) in coating buffer diluted from Coating Solution Concentrate Kit (KPL) at 4°C overnight. Then, the plate was washed with 1× wash buffer (KPL) and blocked with 1% BSA Blocking Solution (KPL) for 1 h at room temperature. Cell supernatant was added to the wells and incubated for 1 h at room temperature in the shaker at 220 rpm. After washing with 1× wash buffer (KPL), each well was incubated with the second anti-M-CSF antibody (1:300, Cat#sc-13103, Santa Cruz Biotech) overnight at 4°C. The wells were washed four times, 5 min for each and incubated with 100 μL of HRP-conjugated goat-anti-rabbit IgG (1:5000, Cat# 111-035-045, Jackson Laboratory) for 1 h at room temperature. The wells were washed four times, 5 min for each and incubated with 100 μL of ABTS ELISA HRP Substrate (KPL). Absorbance at 410 nm was measured by Synergy HT microplate reader (BioTek).
Flow cytometry
MyC-CaP cells were coculured with or without RAW cells, docetaxel (IC10 or IC50) for 48 h before cells were trypsinized. Single cell suspension was rinsed with PBS twice and incubated with APC conjugated anti-IL-10 antibody (Cat#17-7101-82, eBiosicence) for 30 min at 4°C at darkness. Cell acquisition was done on a BD LSR-II flow cytometer (Beckman Coulter) and data were analyzed by FlowJo software (TreeStar).
For tumor tissue analysis, single cell suspension was prepared by digestion of collagenase II at 0.1% for 1 h. Then, cells were counted and incubated with APC-conjugated anti-CD11b antibody (Cat#17-0112-81, eBioscience) and PE-conjugated anti-CSF1R antibody (Cat#12-1152-82, eBioscience) for 30 min at 4°C in darkness.
MyC-Cap subcutaneous xenograft model
All animal experiments were approved by the Animal Research Committee of the University of California, Los Angeles. For MyC-CaP s.c. xenograft model, 16 FVB male mice that are 6–8 weeks old from Taconic Biosciences were adopted and kept at BSL2 animal facility. After trypsinization and rinsing with cooled PBS, 1.0 × 106 MyC-CaP cells were resuspended in 200 μL PBS/Matrigel (1:2) (356230, Corning) and injected with insulin syringe into the subcutaneous space on the right back of FVB male mice (n = 16). One week after the cell injection, mice were castrated and divided randomly into four groups, receiving DMSO vehicle + control chow, DMSO vehicle + chow containing PLX3397, docetaxel + control chow or docetaxel + chow containing PLX3397. The PLX3397 dosage is 40 mg/kg/day on average and docetaxel dosage is 40 mg/kg/week. Tumor size was measured by digital calipers and calculated by the formula V = 0.5 * a * b 2, in which a is the larger and b is the smaller index of the two perpendicular indexes of the tumor.
CWR22Rv1 orthotopic xenograft model
After trypsinization and rinsing with precooled PBS, 1 × 105 CWR22Rv1 cells, stably expressing firefly luciferase, were resuspended in 10 μL of PBS/Matrigel (1:2) (356230, Corning) and injected by insulin syringe into the left anterior lobe of prostate gland of 6–8 weeks old SCID-beige male mice (Jackson Laboratory). All mice were castrated on day 14 post injection and randomly divided into two groups, receive docetaxel + control chow or docetaxel + PLX3397 chow (40 mg/kg/day). The docetaxel treatment started on day 19 at 10 mg/kg/week. The in vivo BLI were performed every week and the luminescence count was recorded as previously described (Palmeri et al. 2008). All mice were killed on day 42.
Statistical analysis
Data are all presented as mean ± s.e.m. Student t-test was used for comparison between two groups while two-way ANOVA was used for comparisons between multiple groups.
Results
Docetaxel-mediated tumor cell injury induces the expression of M2 cytokines
To study the impact of docetaxel in prostate cancer, we first examined the dose response of this chemotherapeutic agent on several prostate cancer cell lines (Fig. 1A). We found that PCa cells exhibit a wide range of sensitivity to docetaxel, with C4-2 (IC50 = 2 nM) and CWR22Rv1 (IC50 = 5 nM) being the most sensitive, PC-3 (IC50 = 30 nM) as an intermediate responder and MyC-CaP (IC50 = 100 nM) being the most resistant. As we have shown in previous studies, conventional cytotoxic therapies such as radiation therapy and ADT all can induce PCa cells to express M2 cytokines (Xu et al. 2013, Escamilla et al. 2015). Here, we further inquired whether docetaxel in addition to ADT would also induce the expression of M2 cytokines such as CSF-1 and IL-10. To mimic ADT, all prostate cancer cells were cultured in media supplemented with charcoal-treated fetal bovine serum (FBS) to remove the androgens. As shown in Fig. 1B MyC-CaP or CWR22Rv1 cells treated with ADT plus docetaxel, dosed at each line’s respective IC50, increased the expression of CSF-1 and IL-10. Likewise, the expression of these M2 cytokines was also induced when PCa cells, including MyC-CaP, CWR22Rv1, PC3 and C4-2, were treated in the presence of macrophages (Fig. 1C). This ADT plus docetaxel treatment resulted in a significant reduction in cell proliferation, as indicated by the decrease in the proliferative marker Ki67. Interestingly, when the PCa cells were treated with a lower dose of docetaxel at the IC10 dose, the elevation of M2 cytokine expression was no longer observed (Fig. 1D). Docetaxel treatment induced increase in CSF-1 and IL-10 in the tumor cells were further analyzed and verified at the protein level by CSF-1 ELISA (Fig. 1E) and IL-10 flow cytometry (Fig. 1F). This induction of M2 cytokines is likely not restricted to docetaxel alone. We observed very similar effects with paclitaxel treatment of all four PCa cell lines (Supplementary Fig. 1). Collectively, these findings support that cell injury mediated by ADT plus docetaxel induces the heightened expression of M2 cytokines in PCa cells.
Docetaxel induces CSF-1 expression and increases the recruitment of macrophages in vitro
CSF-1 or M-CSF is a cytokine critical not only in the differentiation and proliferation of myeloid cells but also in the recruitment and polarization of protumorigenic M2 macrophages (Brown et al. 2017). Next, we examine the impact of macrophage recruitment in the setting of docetaxel treatment. As shown in Fig. 2A and B, CWR22Rv1 and C4-2 PCa cells treated with docetaxel were able to recruit more macrophages in an in vitro transwell assay compared to chemo-naïve cells. The elevated CSF-1 produced by the docetaxel-treated PCa cells likely contributed to the increased macrophage recruitment, as the addition of the CSF-1Ri PLX3397 attenuated the enhancement in macrophage recruitment in vitro (Fig. 2A and B), as we and others have previously reported (Xu et al. 2013, Escamilla et al. 2015, Moughon et al. 2015, Butowski et al. 2016).
PLX3397 is known to also inhibit c-Kit (Tap et al. 2015). We employed a second highly selective CSF-1R kinase inhibitor GW2580 to substantiate that CSF1/CSF1R as the key signal axis for macrophage recruitment (Priceman et al. 2010). As shown in Fig. 2C and D, the enhancement of macrophage recruitment across a transwell mediated by docetaxel-treated PCa cells was dampened significantly by the addition of GW2580.
Adding CSF-1R kinase inhibitor, PLX3397, to docetaxel regimen enhances therapeutic efficacy in CRPC
Next, we investigated the impact of docetaxel treatment on macrophage recruitment in vivo in CRPC tumors. We first evaluated TAMs in the MyC-CaP tumors engrafted subcutaneously in syngeneic FVB male mice. One week after tumor cell implantation, tumor-bearing mice were treated with surgical castration as ADT, and divided into four treatment groups receiving (i) diluent control, (ii) oral PLX3397, (iii) docetaxel or (iv) docetaxel plus PLX3397. The PLX3397 treatment was administered orally via rodent chow and docetaxel was administered IP at 40 mg/kg/week. Comparing to diluent control=treated tumors, PLX3397 only treatment significantly reduced the number of CD11b+ CSF1R+ TAMs, while docetaxel significantly increased TAMs (Fig. 3A and B). Importantly, the addition of PLX3397 to docetaxel-treated group was able to not only reverse the chemotherapy-induced TAM influx but suppressed the TAM level in the tumor below that of the control treated group (Fig. 3A and B). These results demonstrate the importance of CSF-1/CSF-1R axis in the recruitment of macrophages and the effectiveness of PLX3397 in blocking this CSF-1R-mediated TAM recruitment in vitro and in vivo.
In our previous therapeutic studies, we consistently observed that CSF-1R blockade treatment alone can reduce the infiltration of TAMs but exert negligible impact on tumor growth in vivo (Priceman et al. 2010, Xu et al. 2013, Escamilla et al. 2015, Butowski et al. 2016). The same result was observed here in the MyC-CaP tumors: no significant reduction in tumor growth was observed after oral PLX3397 treatment alone despite clear reduction in the level of TAMs in the tumor (Fig. 3C, D and E). As expected, docetaxel treatment significantly retarded the growth of MyC-CaP tumor compared to control (Fig. 3C, D and E). More importantly, docetaxel plus PLX3397 achieved the most significant tumor growth suppression in the four treatment groups, more effective than docetaxel alone (Fig. 3C, D and E).
Next, we asked whether the benefit of PLX3397 in combination with docetaxel in the subcutaneous MyC-CaP model can also be observed in the orthotopic prostatic environment of the CWR22Rv1 model. SCID/Beige male mice received intraprostatic injection of firefly luciferase-labeled CWR22Rv1 cells, such that tumor growth can be monitored in real time by bioluminescence imaging (BLI, Fig. 3F and G). On day 14 after tumor cell implantation, mice received ADT via surgical castration. On day 19, tumor-bearing mice received either docetaxel with control or docetaxel plus oral PLX3397 (Fig. 3B). Treatment continued to day 42, at which point the animals were killed. Assessed either by BLI (Fig. 3F and G) or by terminal tumor volume (Fig. 3H and I), the docetaxel plus PLX3397 group consistently showed significantly greater efficiency in suppressing tumor growth over docetaxel treatment alone. Again, corroborating our prior findings, the added oral PLX3397 drastically reduced the level of CD11b+ CSF1R+ TAMs from 10.6% in the docetaxel only group to 0.1% in the docetaxel plus PLX3397 group, as analyzed by flow cytometry (Fig. 3J). This finding was further verified by F4/80 immunohistochemistry stain to detect macrophages (Fig. 3K). The functional consequences of TAM inhibition by PLX3397 included lowering angiogenic drive, tissue remodeling and immunosuppression as assessed by VEGF-A, MMP-9 and Arg-1 expression respectively (Fig. 3F and G). Taken together, we have shown that the use of a selective CSF-1Ri PLX3397 can block the infiltration of TAMs into prostate tumor and thus reduce the protumorigenic influences of M2 macrophages by lowering tumoral angiogenesis, tissue remodeling and immunosuppression leading to more effective treatment response to docetaxel.
Discussion
Docetaxel is a widely used chemotherapeutic agent in treating breast cancer (Palmeri et al. 2008), head and neck cancer (Rapidis et al. 2008) and non-small-cell lung cancer (Fossella 2002). In the Chemohormonal Therapy vs Androgen Ablation Randomized Trial for Extensive Disease (CHAARTED) randomized phase III trial, men with hormone-naive metastatic PCa were randomly assigned to receive docetaxel plus ADT or ADT alone, with nearly 400 men in each arm. In particular, patients who had high-volume disease benefited the most with docetaxel, achieving a very significant prolongation of their median survival by 17 months compared to ADT alone (Azad et al. 2015). Hence, docetaxel is an important therapeutic agent in the armamentarium against CRPC.
In this study, we investigated whether TAMs, an important component of the tumor microenvironment, could influence CRPC’s response to docetaxel. We postulate that cellular damage sustained during docetaxel treatment induces PCa cells to produce cytokines and chemokines that recruit and polarize macrophages to the protumorigenic, alternatively activated M2 subtype (Brown et al. 2017). Congruent with this concept, we observed a significant increase in the expression of M2 cytokines, such as CSF-1 and IL-10 in all four prostate cancer cell lines, MyC-CaP, PC-3, CWR22Rv1 and C4-2, after docetaxel treatment. The elevated CSF-1 led to increased infiltration of macrophages in vitro and TAMs in MyC-CaP and CWR22Rv1 tumors after ADT and docetaxel treatment. We observed that treatment with another chemotherapeutic agent, paclitaxel, also elicited an increase in M2 cytokine expression in PCa, parallel the findings of a comprehensive chemotherapeutic study in preclinical breast cancer (DeNardo et al. 2011). Importantly, these findings support the rational combination of CSF-1Ri with docetaxel to lower the recruitment and M2 polarization of TAMs, which in turn reduce the protumorigenic influences of TAMs and significantly increase the efficacy of tumor growth suppression of ADT and docetaxel treatment (Fig. 4).
As the emergence of resistance to the current therapies is expected, what new and effective therapies will be incorporated to treat CRPC? A second-line taxane, cabazitaxel, was developed to overcome this resistance problem. The effectiveness of docetaxel is limited by its affinity for P-glycoprotein, an ATP-dependent drug efflux pump that decreases the intracellular concentrations of drugs (Bradshaw & Arceci 1998). Cabazitaxel exhibits low affinity for P-glycoprotein and has been shown to be effective in docetaxel-refractory PCa patients (de Bono et al. 2010, Paller & Antonarakis 2011). Although the cancer vaccine Sipuleucel-T was approved for CRPC, current clinical experience suggests this therapy has limited efficacy for aggressive large volume disease (Schellhammer et al. 2013, Mok et al. 2014). New immunotherapeutic strategies for CRPC need further exploration. In this regard, TAMs could have multiple negative influences. For instance, M2 macrophages are well known to impair T-cell responses by depleting essential nutrients through arginase I or by inhibiting T-cell receptor CD3ζchain (Rodriguez et al. 2004, Munder et al. 2006). Interestingly, a recent study by Gordon et al. (2017) further implicated that PD-1 expressing TAMs are inhibiting tumor immunity, which might further empower the efficacy of the PD-1 or PD-L1 checkpoint blockade.
In our collective experience of studying TAM’s influences in cancer therapy, we observed that TAMs contribute to every stage of PCa progression and therapy. From the control of local disease by radiation therapy (Xu et al. 2013), to the implementation of ADT for more advanced disease (Escamilla et al. 2015), to the use of docetaxel in recurrent CRPC studied here, blocking TAMs with CSF-1Ri in conjunction with these conventional therapies consistently improved therapeutic outcome by prolonging the duration of tumor growth suppression. Of note, the use of CSF-1Ri alone has no therapeutic impact in numerous preclinical models we have studied, including PCa, melanoma and lung cancer (Priceman et al. 2010). A large volume of literature shows that macrophages are educated and polarized by the tumor microenvironment towards the protumorigenic M2 subtype (Brown et al. 2017). We deduced that in the face of cellular injuries induces by conventional therapies, tumor cells secrete a higher level of M2 cytokines and chemokines such as CSF-1, CCL2 and IL10 that accentuate the protumorigenic functions of TAMs. Thus, combining CSF-1Ri with conventional cytotoxic therapies is a rational approach to improve their effectiveness. As we have shown that CSR-1Ri can improve the efficacy of adoptive T-cell therapy (Mok et al. 2014), it will be prudent to consider the incorporation of TAM blockade in combination for future immunotherapy strategies developed for CRPC, be it checkpoint inhibition or CAR T-cell therapy or others (Bilusic et al. 2017). Given the critical role of TAMs in therapeutic setting for PCa, we envision that the incorporation of TAM blockade could extend the efficacy of all phases of treatment. In doing so, we could extend the survival of PCa patients and achieve the goal of transforming PCa into a chronic and survivable malignancy.
Supplementary data
This is linked to the online version of the paper at https://doi.org/10.1530/ERC-18-0284.
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 project was supported by the CDMRP PCRP award W81XWH12-1-0206 and W81XWH 15-1-0256 to L Wu. Flow cytometry and tissue process/immunohistochemical analyses were performed by Flow Cytometry Core Facility and Translational Pathology Core Laboratory of UCLA Jonsson Comprehensive Cancer Center, and UCLA CTSI (NIH P50CA16042 and UL1TR001881). Through a sponsored research agreement, Plexxikon Co. supported this investigation by kindly providing the PLX3397.
References
Antonarakis ES, Lu C, Wang H, Luber B, Nakazawa M, Roeser JC, Chen Y, Mohammad TA, Chen Y, Fedor HL, et al. 2014 AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. New England Journal of Medicine 371 1028–1038. (https://doi.org/10.1056/NEJMoa1315815)
Azad AA, Volik SV, Wyatt AW, Haegert A, Le Bihan S, Bell RH, Anderson SA, McConeghy B, Shukin R, Bazov J, et al. 2015 Androgen receptor gene aberrations in circulating cell-free DNA: biomarkers of therapeutic resistance in castration-resistant prostate cancer. Clin Cancer Research 21 2315–2324. (https://doi.org/10.1158/1078-0432.CCR-14-2666)
Bilusic M, Madan RA & Gulley JL 2017 Immunotherapy of prostate cancer: facts and hopes. Clin Cancer Research 23 6764–6770. (https://doi.org/10.1158/1078-0432.CCR-17-0019)
Bradshaw DM & Arceci RJ 1998 Clinical relevance of transmembrane drug efflux as a mechanism of multidrug resistance. Journal of Clinical Oncology 16 3674–3690. (https://doi.org/10.1200/JCO.1998.16.11.3674)
Brown JM, Recht L & Strober S 2017 The promise of targeting macrophages in cancer therapy. Clinical Cancer Research 23 3241–3250. (https://doi.org/10.1158/1078-0432.CCR-16-3122)
Butowski N, Colman H, De Groot JF, Omuro AM, Nayak L, Wen PY, Cloughesy TF, Marimuthu A, Haidar S, Perry A, et al. 2016 Orally administered colony stimulating factor 1 receptor inhibitor PLX3397 in recurrent glioblastoma: an Ivy Foundation Early Phase Clinical Trials Consortium phase II study. Neuro-Oncology 18 557–564. (https://doi.org/10.1093/neuonc/nov245)
de Bono JS, Oudard S, Ozguroglu M, Hansen S, Machiels JP, Kocak I, Gravis G, Bodrogi I, Mackenzie MJ, Shen L, et al. 2010 Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet 376 1147–1154. (https://doi.org/10.1016/S0140-6736(10)61389-X)
de Bono JS, Logothetis CJ, Molina A, Fizazi K, North S, Chu L, Chi KN, Jones RJ, Goodman OBJ, Saad F, et al. 2011 Abiraterone and increased survival in metastatic prostate cancer. New England Journal of Medicine 364 1995–2005. (https://doi.org/10.1056/NEJMoa1014618)
DeNardo DG, Brennan DJ, Rexhepaj E, Ruffell B, Shiao SL, Madden SF, Gallagher WM, Wadhwani N, Keil SD, Junaid SA, et al. 2011 Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discovery 1 54–67. (https://doi.org/10.1158/2159-8274.CD-10-0028)
Escamilla J, Schokrpur S, Liu C, Priceman SJ, Moughon D, Jiang Z, Pouliot F, Magyar C, Sung JL, Xu J, et al. 2015 CSF1 receptor targeting in prostate cancer reverses macrophage-mediated resistance to androgen blockade therapy. Cancer Research 75 950–962. (https://doi.org/10.1158/0008-5472.CAN-14-0992)
Esch L, Schulz WA & Albers P 2014 Sequential treatment with taxanes and novel anti-androgenic compounds in castration-resistant prostate cancer. Oncology Research and Treatment 37 492–498. (https://doi.org/10.1159/000365530)
Fossella FV 2002 Docetaxel in second-line treatment of non-small-cell lung cancer. Clinical Lung Cancer 3 (Supplement 2) S23–S28. (https://doi.org/10.3816/CLC.2002.s.010)
Gan L, Chen S, Wang Y, Watahiki A, Bohrer L, Sun Z, Wang Y & Huang H 2009 Inhibition of the androgen receptor as a novel mechanism of taxol chemotherapy in prostate cancer. Cancer Research 69 8386–8394. (https://doi.org/10.1158/0008-5472.CAN-09-1504)
Gordon SR, Maute RL, Dulken BW, Hutter G, George BM, McCracken MN, Gupta R, Tsai JM, Sinha R, Corey D, et al. 2017 PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature 545 495–499. (https://doi.org/10.1038/nature22396)
Merseburger AS, Hammerer P, Rozet F, Roumeguère T, Caffo O, da Silva FC & Alcaraz A 2015 Androgen deprivation therapy in castrate-resistant prostate cancer: how important is GnRH agonist backbone therapy? World Journal of Urology 33 1079–1085. (https://doi.org/10.1007/s00345-014-1406-2)
Mok S, Koya RC, Tsui C, Xu J, Robert L, Wu L, Graeber T, West BL, Bollag G & Ribas A 2014 Inhibition of CSF-1 receptor improves the antitumor efficacy of adoptive cell transfer immunotherapy. Cancer Research 74 153–161. (https://doi.org/10.1158/0008-5472.CAN-13-1816)
Moughon DL, He H, Schokrpur S, Jiang ZK, Yaqoob M, David J, Lin C, Iruela-Arispe ML, Dorigo O & Wu L 2015 Macrophage blockade using CSF1R inhibitors reverses the vascular leakage underlying malignant ascites in late-stage epithelial ovarian cancer. Cancer Research 75 4742–4752. (https://doi.org/10.1158/0008-5472.CAN-14-3373)
Munder M, Schneider H, Luckner C, Giese T, Langhans C-D, Fuentes JM, Kropf P, Mueller I, Kolb A, Modolell M, et al. 2006 Suppression of T-cell functions by human granulocyte arginase. Blood 108 1627–1634. (https://doi.org/10.1182/blood-2006-11-010389)
Paller CJ & Antonarakis ES 2011 Cabazitaxel: a novel second-line treatment for metastatic castration-resistant prostate cancer. Drug Design, Development and Therapy 5 117–124. (https://doi.org/10.2147/DDDT.S13029)
Palmeri L, Vaglica M & Palmeri S 2008 Weekly docetaxel in the treatment of metastatic breast cancer. Therapeutics and Clinical Risk Management 4 1047–1059. (https://doi.org/10.2147/TCRM.S3397)
Petrylak DP 2003 Docetaxel for the treatment of hormone-refractory prostate cancer. Reviews in Urology 5 S14–S21.
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. (https://doi.org/10.1056/NEJMoa041318)
Priceman SJ, Sung JL, Shaposhnik Z, Burton JB, Torres-Collado AX, Moughon DL, Johnson M, Lusis AJ, Cohen DA, Iruela-Arispe ML, et al. 2010 Targeting distinct tumor-infiltrating myeloid cells by inhibiting CSF-1 receptor: combating tumor evasion of antiangiogenic therapy. Blood 115 1461–1471. (https://doi.org/10.1182/blood-2009-08-237412)
Rapidis A, Sarlis N, Lefebvre J-L & Kies M 2008 Docetaxel in the treatment of squamous cell carcinoma of the head and neck. Therapeutics and Clinical Risk Management 4 865–886. (https://doi.org/10.2147/TCRM.S3133)
Rodriguez PC, Quiceno DG, Zabaleta J, Ortiz B, Zea AH, Piazuelo MB, Delgado A, Correa P, Brayer J, Sotomayor EM, et al. 2004 Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Research 64 5839–5849. (https://doi.org/10.1158/0008-5472.CAN-04-0465)
Romanel A, Gasi Tandefelt D, Conteduca V, Jayaram A, Casiraghi N, Wetterskog D, Salvi S, Amadori D, Zafeiriou Z, Rescigno P, et al. 2015 Plasma AR and abiraterone-resistant prostate cancer. Science Translational Medicine 7 312re310. (https://doi.org/10.1126/scitranslmed.aac9511)
Ryan CJ, Smith MR, Fizazi K, Saad F, Mulders PF, Sternberg CN, Miller K, Logothetis CJ, Shore ND, Small EJ, et al. 2015 Abiraterone acetate plus prednisone versus placebo plus prednisone in chemotherapy-naive men with metastatic castration-resistant prostate cancer (COU-AA-302): final overall survival analysis of a randomised, double-blind, placebo-controlled phase 3 study. Lancet Oncology 16 152–160. (https://doi.org/10.1016/S1470-2045(14)71205-7)
Schellhammer PF, Chodak G, Whitmore JB, Sims R, Frohlich MW & Kantoff PW 2013 Lower baseline prostate-specific antigen is associated with a greater overall survival benefit from sipuleucel-T in the Immunotherapy for Prostate Adenocarcinoma Treatment (IMPACT) trial. Urology 81 1297–1302. (https://doi.org/10.1016/j.urology.2013.01.061)
Scher HI, Fizazi K, Saad F, Taplin M-E, Sternberg CN, Miller K, de Wit R, Mulders P, Chi KN, Shore ND, et al. 2012 Increased survival with enzalutamide in prostate cancer after chemotherapy. New England Journal of Medicine 367 1187–1197. (https://doi.org/10.1056/NEJMoa1207506)
Siegel RL, Miller KD & Jemal A 2017 Cancer statistics, 2017. CA: A Cancer Journal for Clinicians 67 7–30. (https://doi.org/10.3322/caac.21387)
Silberstein JL, Taylor MN & Antonarakis ES 2016 Novel insights into molecular indicators of response and resistance to modern androgen-axis therapies in prostate cancer. Current Urology Reports 17 29. (https://doi.org/10.1007/s11934-016-0584-4)
Sweeney CJ, Chen Y-H, Carducci M, Liu G, Jarrard DF, Eisenberger M, Wong Y-N, Hahn N, Kohli M, Cooney MM, et al. 2015 Chemohormonal therapy in metastatic hormone-sensitive prostate cancer. New England Journal of Medicine 373 737–746. (https://doi.org/10.1056/NEJMoa1503747)
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. (https://doi.org/10.1056/NEJMoa040720)
Tap WD, Wainberg ZA, Anthony SP, Ibrahim PN, Zhang C, Healey JH, Chmielowski B, Staddon AP, Cohn AL, Shapiro GI, et al. 2015 Structure-guided blockade of CSF1R kinase in tenosynovial giant-cell tumor. New England Journal of Medicine 373 428–437. (https://doi.org/10.1056/NEJMoa1411366)
Xu J, Escamilla J, Mok S, David J, Priceman S, West B, Bollag G, McBride W & Wu L 2013 CSF1R signaling blockade stanches tumor-infiltrating myeloid cells and improves the efficacy of radiotherapy in prostate cancer. Cancer Research 73 2782–2794. (https://doi.org/10.1158/0008-5472.CAN-12-3981)